Fasting-mimicking diet cycles reduce neuroinflammation to attenuate cognitive decline in Alzheimer's models

Priya Rangan, Fleur Lobo, Edoardo Parrella, Nicolas Rochette, Marco Morselli, Terri-Leigh Stephen, Anna Laura Cremonini, Luca Tagliafico, Angelica Persia, Irene Caffa, Fiammetta Monacelli, Patrizio Odetti, Tommaso Bonfiglio, Alessio Nencioni, Martina Pigliautile, Virginia Boccardi, Patrizia Mecocci, Christian J Pike, Pinchas Cohen, Mary Jo LaDu, Matteo Pellegrini, Kyle Xia, Katelynn Tran, Brandon Ann, Dolly Chowdhury, Valter D Longo, Priya Rangan, Fleur Lobo, Edoardo Parrella, Nicolas Rochette, Marco Morselli, Terri-Leigh Stephen, Anna Laura Cremonini, Luca Tagliafico, Angelica Persia, Irene Caffa, Fiammetta Monacelli, Patrizio Odetti, Tommaso Bonfiglio, Alessio Nencioni, Martina Pigliautile, Virginia Boccardi, Patrizia Mecocci, Christian J Pike, Pinchas Cohen, Mary Jo LaDu, Matteo Pellegrini, Kyle Xia, Katelynn Tran, Brandon Ann, Dolly Chowdhury, Valter D Longo

Abstract

The effects of fasting-mimicking diet (FMD) cycles in reducing many aging and disease risk factors indicate it could affect Alzheimer's disease (AD). Here, we show that FMD cycles reduce cognitive decline and AD pathology in E4FAD and 3xTg AD mouse models, with effects superior to those caused by protein restriction cycles. In 3xTg mice, long-term FMD cycles reduce hippocampal Aβ load and hyperphosphorylated tau, enhance genesis of neural stem cells, decrease microglia number, and reduce expression of neuroinflammatory genes, including superoxide-generating NADPH oxidase (Nox2). 3xTg mice lacking Nox2 or mice treated with the NADPH oxidase inhibitor apocynin also display improved cognition and reduced microglia activation compared with controls. Clinical data indicate that FMD cycles are feasible and generally safe in a small group of AD patients. These results indicate that FMD cycles delay cognitive decline in AD models in part by reducing neuroinflammation and/or superoxide production in the brain.

Trial registration: ClinicalTrials.gov NCT05480358.

Keywords: Alzheimer’s disease; CP: Metabolism; CP: Neuroscience; NADPH oxidase; amyloid beta; fasting; fasting-mimicking diet; hyperphosphorylated tau; microglia; neuroinflammation; protein restriction; superoxide.

Conflict of interest statement

Declaration of interests V.D.L. has equity interest in L-Nutra, which develops and sells medical food for the prevention and treatment of diseases. A patent on this work has been filed by the University of Southern California, which has equity interest in L-Nutra (Fasting-mimicking Diet (FMD) as an Intervention for Alzheimer’s Disease (AD), US 62/840,762, Filed Apr 15, 2019).

Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.. FMD cycles improve cognitive behavior…
Figure 1.. FMD cycles improve cognitive behavior in female E4FAD mice
(A) Experimental diet and behavior schedule for female E4FAD mice starting at 2.5 months of age through 7–7.5 months of age. (B) SAB percentage for E4FAD females at baseline (2.5 months) and at 6 or 6.5 months after ~3 months of diet (baseline, n = 19; control, 6 months, n = 9; FMD, 6 months, n = 11). (C) Latency (seconds lapsed before finding escape box) between E4FAD FMD females (n = 20) and E4FAD females on control diet (n = 19) in the Barnes maze at approximately 6.5–7 months. (D) Success rate in finding the escape box between E4FAD FMD females (n = 20) and E4FAD females on control diet (n = 19) in the Barnes maze at approximately 6.5–7 months. (E) Strategies (random, serial, and spatial) used by female E4FAD control group (n = 19) to locate escape box. (F) Strategies (random, serial, and spatial) used by female E4FAD FMD group (n = 20) to locate escape box. Data are presented as mean ± SEM. *p

Figure 2.. FMD cycles reduce hippocampal and…

Figure 2.. FMD cycles reduce hippocampal and cortex Aβ load, Aβ peptides, and neuroinflammatory cytokines,…

Figure 2.. FMD cycles reduce hippocampal and cortex Aβ load, Aβ peptides, and neuroinflammatory cytokines, while increasing hippocampal neurogenesis markers in E4FAD mice
(A) Representative images showing Aβ immunoreactivity in subiculum and cortical regions of female E4FAD control and FMD groups. (B) Quantification of subiculum Aβ load (%) for female E4FAD control (n = 18) and FMD (n = 18) groups. (C) Quantification of CA1 Aβ load (%) for female E4FAD control (n = 18) and FMD (n = 18) groups. (D) Quantification of cortex Aβ load (%) for female E4FAD control (n = 17) and FMD (n = 17) groups. (E) Quantification of triton-soluble Aβ38 for female E4FAD control (n = 7) and FMD (n = 7) groups. (F) Quantification of triton-soluble Aβ40 for female E4FAD control (n = 7) and FMD (n = 7) groups. (G) Quantification of triton-soluble Aβ42 for female E4FAD control (n = 7) and FMD (n = 7) groups. (H) Representative images showing Sox2+, Ki67+, and co-stain hippocampal immunohistochemistry for ~7–7.5-month-old female E4FAD control and FMD groups. White arrows indicate the Ki67+/Sox2+ foci. (I) Quantification of ~7–7.5-month-old E4FAD female Sox2+ (control [n = 13] and FMD [n = 11]) cells in the dentate gyrus (DG) after ~4 months of FMD cycles. (J) Quantification of ~7–7.5-month-old E4FAD female Ki67+Sox2+ (control [n = 13] and FMD [n = 11]) cells in the DG after ~4 months of FMD cycles. (K) Quantification of IL-2p70 in TBS-soluble cortex extract in control (n = 7) and FMD (n = 7) ~7–7.5-month-old female E4FAD mice. (L) Quantification of IL-2 in TBS-soluble cortex extract in control (n = 6) and FMD (n = 7) ~7–7.5-month-old female E4FAD mice. (M) Quantification of TNFα in detergent-soluble/triton-soluble cortex extract in control (n = 7) and FMD (n = 7) ~7–7.5-month-old female E4FAD mice. Data are presented as mean ± SEM. *p

Figure 3.. FMD cycles improve cognitive behavior…

Figure 3.. FMD cycles improve cognitive behavior in 3xTg mice

(A) Experimental diet and behavior…

Figure 3.. FMD cycles improve cognitive behavior in 3xTg mice
(A) Experimental diet and behavior schedule for 3xTg males and females starting at 3.5 months of age through 18.5 months of age. (B) Kaplan-Meier survival curves for 3xTg females in control (n = 25/28), FMD (n = 16/17), and 4% PR (n = 14/16). (C) Kaplan-Meier survival curves for 3xTg males in control (n = 17/18), FMD (n = 16/17), and 4% PR (n = 14/16) groups. (D) SAB score (%) for 8.5-month-old C57B/6 WT females (n = 8), 10.5-month-old 3xTg female control (n = 14), FMD (n = 14), and 4% PR (n = 16) groups. (E) SAB score (%) for 8.5-month-old C57B/6 WT males (n = 8), 10.5-month-old 3xTg male control (n = 13), FMD (n = 16), and 4% PR (n = 15) groups. (F) Recognition index (RI) scored as a percentage for trial 2 (time in seconds spent exploring novel object versus old object) of NOR task for 8.5-month-old C57B/6 WT females (n = 7), and 10.5-month-old 3xTg female control (n = 14), FMD (n = 13), and 4% PR (n = 13) groups. (G) RI for trial 2 of NOR task for 8.5-month-old C57B/6 WT males (n = 8), and 10.5-month-old 3xTg male control (n = 14), FMD (n = 14), and 4% PR (n = 16) groups. Figures 1B and 1C: n = mice that survived to 18 months of age/total mice enrolled in study for group. Figures 1D–1G: 8.5-month-old C57B/6 WT male and female data shown here are the same data as from Figure 5 and S3 and are shown as a comparison with aging 3xTg mice with or without previous treatment cycles. Data are presented as mean ± SEM. *p

Figure 4.. FMD cycles slow the progression…

Figure 4.. FMD cycles slow the progression of AD-associated pathology, increase levels of hippocampal neurogenesis…

Figure 4.. FMD cycles slow the progression of AD-associated pathology, increase levels of hippocampal neurogenesis markers, and regulate microglia levels and activation in aged 3xTg mice
(A) Representative images showing Aβ immunoreactivity and AT8-positive neurons (recognizes abnormally phosphorylated tau) in subiculum or CA1 hippocampus regions of female control, FMD, and 4% PR 3xTg mice. (B) Quantification of subiculum Aβ load (%) for female 3xTg control (n = 21), FMD (n = 16), and 4% PR (n = 14) mice. (C) Quantification of CA1 Aβ load (%) for female 3xTg control (n = 20), FMD (n = 16), and 4% PR (n = 14) mice. (D) Quantification of AT8+ neurons in the subiculum and CA1 for female 3xTg control (n = 20), FMD (n = 14), and PR (n = 12) mice. (E) Representative images showing Aβ immunoreactivity and AT8+ neurons in subiculum or CA1 hippocampus regions of male 3xTg control, FMD, and 4% PR mice. (F) Quantification of subiculum Aβ load (%) for male 3xTg control (n = 15), FMD (n = 14), and 4%PR (n = 12) mice. (G) Quantification of CA1 Aβ load (%) for male 3xTg control (n = 15), FMD (n = 14), and 4%PR (n = 12) mice. (H) Quantification of AT8+ neurons in the subiculum and CA1 for male 3xTg control (n = 12), FMD (n = 13), and 4%PR (n = 11) mice. (I) Representative images showing BrdU+ DAB-immunohistochemistry in DG of hippocampus for 18.5-month-old female 3xTg control and FMD groups (left). Quantification of BrdU+ cells within the SGZ and inner third of the granule cell layer of the DG for 18.5-month-old female 3xTg control (n = 15) and FMD (n = 15) groups (right). (J) Representative images showing Sox2+, BrdU+, and co-stain hippocampal immunohistochemistry for 18.5-month-old female 3xTg control and FMD groups (left). Quantification of 18.5-month-old 3xTg female BrdU+Sox2+ (control [n = 9] and FMD [n = 8]) cells within the SGZ and inner third of the granule cell layer of the DG after ~15 months of FMD cycles (right). (K) Representative images showing CD11b-ir microglia in hippocampus sections of 18.5-month-old female C57B/6 WT, 3xTg control, and 3xTg FMD mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of C57B/6 WT, 3xTg control, and 3xTg FMD groups (bottom left; n = 5–7 animals per group). Percentage of different microglia activation stages (from 1 to 4) of C57B/6 WT, 3xTg control, and 3xTg FMD mice (bottom right; n = 5–7 animals per group). (L) Representative images showing BrdU+ DAB-immunohistochemistry in DG of hippocampus for 18.5-month-old male 3xTg control and FMD groups (left). Quantification of BrdU+ cells within the SGZ and inner third of the granule cell layer of the DG for 18.5-month-old male 3xTg control (n = 10) and FMD (n = 10) groups (right). (M) Representative images showing Sox2+, BrdU+, and co-stain hippocampal immunohistochemistry for 18.5-month-old male 3xTg control and FMD groups (left). Quantification of 18.5-month-old 3xTg male BrdU+Sox2+ (control [n = 11] and FMD [n = 9]) cells within the SGZ and inner third of the granule cell layer of the DG after ~15 months of FMD cycles (right). (N) Representative images showing CD11b-ir microglia in hippocampus sections of 18.5-month-old male C57B/6 WT, 3xTg control, and 3xTg FMD mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of C57B/6 WT, 3xTg control, and 3xTg FMD (bottom left; n = 5–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of C57B/6 WT, 3xTg control, and 3xTg FMD mice (bottom right; n = 5–8 animals per group). (O) Representative images showing CD68+ microglia in hippocampus sections of 18.5-month-old male 3xTg control and 3xTg FMD mice (top; n = 2–3 animals per group). Quantification of density of CD68+ cells in hippocampus CA1 and subiculum combined brain regions of 3xTg control and 3xTg FMD (bottom; n = 2–3 animals per group. Data are presented as mean ± SEM. (B–D and F–H) *p

Figure 5.. Short-term treatment with FMD cycles…

Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation,…

Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation, and reduces the expression of neuroinflammation genes, microglial activation, and reduced tau phosphorylation in 3xTg mice
(A) Experimental diet and behavior schedule for 3xTg males and females starting at 6.5 months of age through approximately 8.5 months of age for five FMD cycles. (B) RI for trial 2 of NOR task for 8.5-month-old C57B/6 WT males (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after four cycles of FMD and 7 days of refeeding (n = 5). (C) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg female control and FMD groups. (D) Quantification of subiculum Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (E) Quantification of CA1 Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (F) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg female control (n = 8) and FMD (n = 8) groups. (G) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg male control and FMD groups. (H) Quantification of subiculum Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (I) Quantification of CA1 Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (J) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg male control (n = 21) and FMD (n = 7) groups. (K) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old female C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom right). (L) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old male C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom right). (M) Representative images of confocal stack immune reactive for Iba1 microglia (red) in the prefrontal cortex for male and female 3xTg control diet and FMD cohorts. Scale bars, 100 μm. (N) Example 3D skeletonized microglial projections for 3xTg control diet and 3xTg FMD cohorts. (O and P) Quantification of Iba1 immuno-reactive soma area (O) and circularity (P) in male (M) and female (F) 3×Tg mice with control diet (open bars) and FMD (filled bars) from high-magnification images (n = 2/group). (Q) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old female 3xTg controls (n = 3) and 3xTg females after one 4-day cycle of FMD, with no refeeding (n = 6). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. (R) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old male 3xTg controls (n = 9) and 3xTg males after four cycles of FMD and 2 days of refeeding (n = 9). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. Data are presented as mean ± SEM. (B) *p

Figure 6.. Short-term cycles of FMD regulate…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice,…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice, while NOX2 deletion or inhibition improves cognitive behavior, mitigates pathology progression, and reduces microglia activation
(A) Diagram describing the dual positive and negative function of microglia. Microglia are involved in neuronal development and repair, but their production of O2− into ONOO− can promote toxicity and lead to neurodegeneration. (B) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female 3xTg controls (n = 4) and 3xTg females after one 4-day cycle of FMD with no refeeding (n = 10). (C) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female C57B/6 WT (n = 5) and 8.5-month-old 3xTg female control (n = 6) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (D) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old male C57B/6 WT (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (E) Quantification of Nox2 levels in cortex extract of control (n = 5) and FMD (n = 4) ~7-7.5-month-old female E4FAD mice after 4 months of biweekly FMD cycles (measured as Nox2/Vinculin protein expression levels). (F) Western blot image used for quantifying Nox2 levels in whole-cortex extract of control (n = 5) and FMD (n = 4) ~7- to 7.5-month-old female E4FAD mice. Vinculin was loading control (bottom). (G) 3xTg/Nox2-KO mice generation and experimental design. The experimental design of the tests conducted on 3xTg/Nox2-KO and control mice is depicted, as well as a schematic representation of the breeding strategy used to develop 3xTg/Nox2-KO mice and corresponding WT (mixed background 129/B6/B6). Some mice were euthanized for pathology at 13–14 months, while those used for fear-conditioning (FC) tests were aged until 15–18 months. (H) Representative images showing CD11b-ir microglia in hippocampus sections of 13- to 14-month-old male WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO (bottom left; n = 4–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (bottom right; n = 4–8 animals per group). (I) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice were tested with the Y-maze apparatus (12.5 months of age). SAB scores obtained through Y-maze task are shown (n = 7–23 per group). (J) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) FC tests. Freezing times (%) 24-h post shock for FC are shown (n = 6–22 per group) (left). WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) underwent FC tests. Freezing times (%) 48-h post shock for FC are shown (n = 6–22 per group) (right). (K) Representative images showing AT8 antibody (recognizes abnormally phosphorylated tau) immunoreactivity in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (left). Quantification of total AT8-immuno-reactive cells in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (n = 10–13/group) (right). (L) Experimental design of apocynin treatment. Eight-month-old WT (129/B6 background) and 3xTg mice were treated with apocynin-dissolved drinkable water or apocynin-free water for 6 months. During the final 4 weeks of treatment, the animals were tested using the Y-maze apparatus, and NOR and Rotarod assays. After completion of the behavioral tasks, the mice were euthanized and their brains analyzed. (M) Comparison of SAB scores between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice using the Y-maze apparatus (n = 10–13 animals per group). (N) Comparison of RI values between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin- treated mice during NOR test (n = 10–13 animals per group). (O) Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice (n = 5–10 animals/group). Data are presented as mean ± SEM. (H and O) *p

Figure 7.. Safety and feasibility of FMD…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI or mild AD
(A) A phase I/II randomized and placebo-controlled (single-blind) clinical study for 40 patients with aMCI or mild AD was designed for 12 monthly FMD cycles; 28/40 enrolled patients were randomly assigned to a placebo (control) diet arm (n = 16) or an FMD (ProLonAD) arm (n = 12). In the placebo arm, a gray square indicates that, for 5 days a month, patients are assigned a diet in which lunch or dinner is replaced with a meal based on pasta or rice with vegetables, without added supplements. In the FMD arm, a black square indicates a 5-day FMD cycle, followed by a striped box that indicates 25 days of a normal diet. White box indicates an in-between period between FMD cycles. Primary and secondary endpoints collected at each visit (per FMD cycles completed) is described in lower right-hand corner. (B) Graphical representation of the effects of FMD on the E4FAD and the 3xTg mouse models of AD, through which it mediates AD-associated pathology, neuroinflammation, NSCs, and Nox2 levels.
All figures (7)
Similar articles
References
    1. ‘t Hart BA, Copray S, and Philippens I (2014). Apocynin, a low molecular oral treatment for neurodegenerative disease. BioMed Res. Int, 298020. - PMC - PubMed
    1. Abels ER, Maas SLN, Nieland L, Wei Z, Cheah PS, Tai E, Kolsteeg CJ, Dusoswa SA, Ting DT, Hickman S, et al. (2019). Glioblastoma-associated microglia reprogramming is mediated by functional transfer of extracellular miR-21. Cell Rep. 28, 3105–3119.e7. - PMC - PubMed
    1. Alves S, Churlaud G, Audrain M, Michaelsen-Preusse K, Fol R, Souchet B, Braudeau J, Korte M, Klatzmann D, and Cartier N (2017). Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain 140, 826–842. - PubMed
    1. Anders S, Pyl PT, and Huber W (2015). HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169. - PMC - PubMed
    1. Anderson AJ, Cummings BJ, and Cotman CW (1994). Increased immunoreactivity for Jun- and Fos-related proteins in Alzheimer’s disease: association with pathology. Exp. Neurol 125, 286–295. - PubMed
Show all 135 references
Publication types
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2.. FMD cycles reduce hippocampal and…
Figure 2.. FMD cycles reduce hippocampal and cortex Aβ load, Aβ peptides, and neuroinflammatory cytokines, while increasing hippocampal neurogenesis markers in E4FAD mice
(A) Representative images showing Aβ immunoreactivity in subiculum and cortical regions of female E4FAD control and FMD groups. (B) Quantification of subiculum Aβ load (%) for female E4FAD control (n = 18) and FMD (n = 18) groups. (C) Quantification of CA1 Aβ load (%) for female E4FAD control (n = 18) and FMD (n = 18) groups. (D) Quantification of cortex Aβ load (%) for female E4FAD control (n = 17) and FMD (n = 17) groups. (E) Quantification of triton-soluble Aβ38 for female E4FAD control (n = 7) and FMD (n = 7) groups. (F) Quantification of triton-soluble Aβ40 for female E4FAD control (n = 7) and FMD (n = 7) groups. (G) Quantification of triton-soluble Aβ42 for female E4FAD control (n = 7) and FMD (n = 7) groups. (H) Representative images showing Sox2+, Ki67+, and co-stain hippocampal immunohistochemistry for ~7–7.5-month-old female E4FAD control and FMD groups. White arrows indicate the Ki67+/Sox2+ foci. (I) Quantification of ~7–7.5-month-old E4FAD female Sox2+ (control [n = 13] and FMD [n = 11]) cells in the dentate gyrus (DG) after ~4 months of FMD cycles. (J) Quantification of ~7–7.5-month-old E4FAD female Ki67+Sox2+ (control [n = 13] and FMD [n = 11]) cells in the DG after ~4 months of FMD cycles. (K) Quantification of IL-2p70 in TBS-soluble cortex extract in control (n = 7) and FMD (n = 7) ~7–7.5-month-old female E4FAD mice. (L) Quantification of IL-2 in TBS-soluble cortex extract in control (n = 6) and FMD (n = 7) ~7–7.5-month-old female E4FAD mice. (M) Quantification of TNFα in detergent-soluble/triton-soluble cortex extract in control (n = 7) and FMD (n = 7) ~7–7.5-month-old female E4FAD mice. Data are presented as mean ± SEM. *p

Figure 3.. FMD cycles improve cognitive behavior…

Figure 3.. FMD cycles improve cognitive behavior in 3xTg mice

(A) Experimental diet and behavior…

Figure 3.. FMD cycles improve cognitive behavior in 3xTg mice
(A) Experimental diet and behavior schedule for 3xTg males and females starting at 3.5 months of age through 18.5 months of age. (B) Kaplan-Meier survival curves for 3xTg females in control (n = 25/28), FMD (n = 16/17), and 4% PR (n = 14/16). (C) Kaplan-Meier survival curves for 3xTg males in control (n = 17/18), FMD (n = 16/17), and 4% PR (n = 14/16) groups. (D) SAB score (%) for 8.5-month-old C57B/6 WT females (n = 8), 10.5-month-old 3xTg female control (n = 14), FMD (n = 14), and 4% PR (n = 16) groups. (E) SAB score (%) for 8.5-month-old C57B/6 WT males (n = 8), 10.5-month-old 3xTg male control (n = 13), FMD (n = 16), and 4% PR (n = 15) groups. (F) Recognition index (RI) scored as a percentage for trial 2 (time in seconds spent exploring novel object versus old object) of NOR task for 8.5-month-old C57B/6 WT females (n = 7), and 10.5-month-old 3xTg female control (n = 14), FMD (n = 13), and 4% PR (n = 13) groups. (G) RI for trial 2 of NOR task for 8.5-month-old C57B/6 WT males (n = 8), and 10.5-month-old 3xTg male control (n = 14), FMD (n = 14), and 4% PR (n = 16) groups. Figures 1B and 1C: n = mice that survived to 18 months of age/total mice enrolled in study for group. Figures 1D–1G: 8.5-month-old C57B/6 WT male and female data shown here are the same data as from Figure 5 and S3 and are shown as a comparison with aging 3xTg mice with or without previous treatment cycles. Data are presented as mean ± SEM. *p

Figure 4.. FMD cycles slow the progression…

Figure 4.. FMD cycles slow the progression of AD-associated pathology, increase levels of hippocampal neurogenesis…

Figure 4.. FMD cycles slow the progression of AD-associated pathology, increase levels of hippocampal neurogenesis markers, and regulate microglia levels and activation in aged 3xTg mice
(A) Representative images showing Aβ immunoreactivity and AT8-positive neurons (recognizes abnormally phosphorylated tau) in subiculum or CA1 hippocampus regions of female control, FMD, and 4% PR 3xTg mice. (B) Quantification of subiculum Aβ load (%) for female 3xTg control (n = 21), FMD (n = 16), and 4% PR (n = 14) mice. (C) Quantification of CA1 Aβ load (%) for female 3xTg control (n = 20), FMD (n = 16), and 4% PR (n = 14) mice. (D) Quantification of AT8+ neurons in the subiculum and CA1 for female 3xTg control (n = 20), FMD (n = 14), and PR (n = 12) mice. (E) Representative images showing Aβ immunoreactivity and AT8+ neurons in subiculum or CA1 hippocampus regions of male 3xTg control, FMD, and 4% PR mice. (F) Quantification of subiculum Aβ load (%) for male 3xTg control (n = 15), FMD (n = 14), and 4%PR (n = 12) mice. (G) Quantification of CA1 Aβ load (%) for male 3xTg control (n = 15), FMD (n = 14), and 4%PR (n = 12) mice. (H) Quantification of AT8+ neurons in the subiculum and CA1 for male 3xTg control (n = 12), FMD (n = 13), and 4%PR (n = 11) mice. (I) Representative images showing BrdU+ DAB-immunohistochemistry in DG of hippocampus for 18.5-month-old female 3xTg control and FMD groups (left). Quantification of BrdU+ cells within the SGZ and inner third of the granule cell layer of the DG for 18.5-month-old female 3xTg control (n = 15) and FMD (n = 15) groups (right). (J) Representative images showing Sox2+, BrdU+, and co-stain hippocampal immunohistochemistry for 18.5-month-old female 3xTg control and FMD groups (left). Quantification of 18.5-month-old 3xTg female BrdU+Sox2+ (control [n = 9] and FMD [n = 8]) cells within the SGZ and inner third of the granule cell layer of the DG after ~15 months of FMD cycles (right). (K) Representative images showing CD11b-ir microglia in hippocampus sections of 18.5-month-old female C57B/6 WT, 3xTg control, and 3xTg FMD mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of C57B/6 WT, 3xTg control, and 3xTg FMD groups (bottom left; n = 5–7 animals per group). Percentage of different microglia activation stages (from 1 to 4) of C57B/6 WT, 3xTg control, and 3xTg FMD mice (bottom right; n = 5–7 animals per group). (L) Representative images showing BrdU+ DAB-immunohistochemistry in DG of hippocampus for 18.5-month-old male 3xTg control and FMD groups (left). Quantification of BrdU+ cells within the SGZ and inner third of the granule cell layer of the DG for 18.5-month-old male 3xTg control (n = 10) and FMD (n = 10) groups (right). (M) Representative images showing Sox2+, BrdU+, and co-stain hippocampal immunohistochemistry for 18.5-month-old male 3xTg control and FMD groups (left). Quantification of 18.5-month-old 3xTg male BrdU+Sox2+ (control [n = 11] and FMD [n = 9]) cells within the SGZ and inner third of the granule cell layer of the DG after ~15 months of FMD cycles (right). (N) Representative images showing CD11b-ir microglia in hippocampus sections of 18.5-month-old male C57B/6 WT, 3xTg control, and 3xTg FMD mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of C57B/6 WT, 3xTg control, and 3xTg FMD (bottom left; n = 5–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of C57B/6 WT, 3xTg control, and 3xTg FMD mice (bottom right; n = 5–8 animals per group). (O) Representative images showing CD68+ microglia in hippocampus sections of 18.5-month-old male 3xTg control and 3xTg FMD mice (top; n = 2–3 animals per group). Quantification of density of CD68+ cells in hippocampus CA1 and subiculum combined brain regions of 3xTg control and 3xTg FMD (bottom; n = 2–3 animals per group. Data are presented as mean ± SEM. (B–D and F–H) *p

Figure 5.. Short-term treatment with FMD cycles…

Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation,…

Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation, and reduces the expression of neuroinflammation genes, microglial activation, and reduced tau phosphorylation in 3xTg mice
(A) Experimental diet and behavior schedule for 3xTg males and females starting at 6.5 months of age through approximately 8.5 months of age for five FMD cycles. (B) RI for trial 2 of NOR task for 8.5-month-old C57B/6 WT males (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after four cycles of FMD and 7 days of refeeding (n = 5). (C) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg female control and FMD groups. (D) Quantification of subiculum Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (E) Quantification of CA1 Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (F) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg female control (n = 8) and FMD (n = 8) groups. (G) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg male control and FMD groups. (H) Quantification of subiculum Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (I) Quantification of CA1 Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (J) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg male control (n = 21) and FMD (n = 7) groups. (K) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old female C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom right). (L) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old male C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom right). (M) Representative images of confocal stack immune reactive for Iba1 microglia (red) in the prefrontal cortex for male and female 3xTg control diet and FMD cohorts. Scale bars, 100 μm. (N) Example 3D skeletonized microglial projections for 3xTg control diet and 3xTg FMD cohorts. (O and P) Quantification of Iba1 immuno-reactive soma area (O) and circularity (P) in male (M) and female (F) 3×Tg mice with control diet (open bars) and FMD (filled bars) from high-magnification images (n = 2/group). (Q) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old female 3xTg controls (n = 3) and 3xTg females after one 4-day cycle of FMD, with no refeeding (n = 6). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. (R) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old male 3xTg controls (n = 9) and 3xTg males after four cycles of FMD and 2 days of refeeding (n = 9). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. Data are presented as mean ± SEM. (B) *p

Figure 6.. Short-term cycles of FMD regulate…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice,…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice, while NOX2 deletion or inhibition improves cognitive behavior, mitigates pathology progression, and reduces microglia activation
(A) Diagram describing the dual positive and negative function of microglia. Microglia are involved in neuronal development and repair, but their production of O2− into ONOO− can promote toxicity and lead to neurodegeneration. (B) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female 3xTg controls (n = 4) and 3xTg females after one 4-day cycle of FMD with no refeeding (n = 10). (C) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female C57B/6 WT (n = 5) and 8.5-month-old 3xTg female control (n = 6) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (D) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old male C57B/6 WT (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (E) Quantification of Nox2 levels in cortex extract of control (n = 5) and FMD (n = 4) ~7-7.5-month-old female E4FAD mice after 4 months of biweekly FMD cycles (measured as Nox2/Vinculin protein expression levels). (F) Western blot image used for quantifying Nox2 levels in whole-cortex extract of control (n = 5) and FMD (n = 4) ~7- to 7.5-month-old female E4FAD mice. Vinculin was loading control (bottom). (G) 3xTg/Nox2-KO mice generation and experimental design. The experimental design of the tests conducted on 3xTg/Nox2-KO and control mice is depicted, as well as a schematic representation of the breeding strategy used to develop 3xTg/Nox2-KO mice and corresponding WT (mixed background 129/B6/B6). Some mice were euthanized for pathology at 13–14 months, while those used for fear-conditioning (FC) tests were aged until 15–18 months. (H) Representative images showing CD11b-ir microglia in hippocampus sections of 13- to 14-month-old male WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO (bottom left; n = 4–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (bottom right; n = 4–8 animals per group). (I) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice were tested with the Y-maze apparatus (12.5 months of age). SAB scores obtained through Y-maze task are shown (n = 7–23 per group). (J) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) FC tests. Freezing times (%) 24-h post shock for FC are shown (n = 6–22 per group) (left). WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) underwent FC tests. Freezing times (%) 48-h post shock for FC are shown (n = 6–22 per group) (right). (K) Representative images showing AT8 antibody (recognizes abnormally phosphorylated tau) immunoreactivity in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (left). Quantification of total AT8-immuno-reactive cells in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (n = 10–13/group) (right). (L) Experimental design of apocynin treatment. Eight-month-old WT (129/B6 background) and 3xTg mice were treated with apocynin-dissolved drinkable water or apocynin-free water for 6 months. During the final 4 weeks of treatment, the animals were tested using the Y-maze apparatus, and NOR and Rotarod assays. After completion of the behavioral tasks, the mice were euthanized and their brains analyzed. (M) Comparison of SAB scores between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice using the Y-maze apparatus (n = 10–13 animals per group). (N) Comparison of RI values between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin- treated mice during NOR test (n = 10–13 animals per group). (O) Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice (n = 5–10 animals/group). Data are presented as mean ± SEM. (H and O) *p

Figure 7.. Safety and feasibility of FMD…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI or mild AD
(A) A phase I/II randomized and placebo-controlled (single-blind) clinical study for 40 patients with aMCI or mild AD was designed for 12 monthly FMD cycles; 28/40 enrolled patients were randomly assigned to a placebo (control) diet arm (n = 16) or an FMD (ProLonAD) arm (n = 12). In the placebo arm, a gray square indicates that, for 5 days a month, patients are assigned a diet in which lunch or dinner is replaced with a meal based on pasta or rice with vegetables, without added supplements. In the FMD arm, a black square indicates a 5-day FMD cycle, followed by a striped box that indicates 25 days of a normal diet. White box indicates an in-between period between FMD cycles. Primary and secondary endpoints collected at each visit (per FMD cycles completed) is described in lower right-hand corner. (B) Graphical representation of the effects of FMD on the E4FAD and the 3xTg mouse models of AD, through which it mediates AD-associated pathology, neuroinflammation, NSCs, and Nox2 levels.
All figures (7)
Similar articles
References
    1. ‘t Hart BA, Copray S, and Philippens I (2014). Apocynin, a low molecular oral treatment for neurodegenerative disease. BioMed Res. Int, 298020. - PMC - PubMed
    1. Abels ER, Maas SLN, Nieland L, Wei Z, Cheah PS, Tai E, Kolsteeg CJ, Dusoswa SA, Ting DT, Hickman S, et al. (2019). Glioblastoma-associated microglia reprogramming is mediated by functional transfer of extracellular miR-21. Cell Rep. 28, 3105–3119.e7. - PMC - PubMed
    1. Alves S, Churlaud G, Audrain M, Michaelsen-Preusse K, Fol R, Souchet B, Braudeau J, Korte M, Klatzmann D, and Cartier N (2017). Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain 140, 826–842. - PubMed
    1. Anders S, Pyl PT, and Huber W (2015). HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169. - PMC - PubMed
    1. Anderson AJ, Cummings BJ, and Cotman CW (1994). Increased immunoreactivity for Jun- and Fos-related proteins in Alzheimer’s disease: association with pathology. Exp. Neurol 125, 286–295. - PubMed
Show all 135 references
Publication types
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3.. FMD cycles improve cognitive behavior…
Figure 3.. FMD cycles improve cognitive behavior in 3xTg mice
(A) Experimental diet and behavior schedule for 3xTg males and females starting at 3.5 months of age through 18.5 months of age. (B) Kaplan-Meier survival curves for 3xTg females in control (n = 25/28), FMD (n = 16/17), and 4% PR (n = 14/16). (C) Kaplan-Meier survival curves for 3xTg males in control (n = 17/18), FMD (n = 16/17), and 4% PR (n = 14/16) groups. (D) SAB score (%) for 8.5-month-old C57B/6 WT females (n = 8), 10.5-month-old 3xTg female control (n = 14), FMD (n = 14), and 4% PR (n = 16) groups. (E) SAB score (%) for 8.5-month-old C57B/6 WT males (n = 8), 10.5-month-old 3xTg male control (n = 13), FMD (n = 16), and 4% PR (n = 15) groups. (F) Recognition index (RI) scored as a percentage for trial 2 (time in seconds spent exploring novel object versus old object) of NOR task for 8.5-month-old C57B/6 WT females (n = 7), and 10.5-month-old 3xTg female control (n = 14), FMD (n = 13), and 4% PR (n = 13) groups. (G) RI for trial 2 of NOR task for 8.5-month-old C57B/6 WT males (n = 8), and 10.5-month-old 3xTg male control (n = 14), FMD (n = 14), and 4% PR (n = 16) groups. Figures 1B and 1C: n = mice that survived to 18 months of age/total mice enrolled in study for group. Figures 1D–1G: 8.5-month-old C57B/6 WT male and female data shown here are the same data as from Figure 5 and S3 and are shown as a comparison with aging 3xTg mice with or without previous treatment cycles. Data are presented as mean ± SEM. *p

Figure 4.. FMD cycles slow the progression…

Figure 4.. FMD cycles slow the progression of AD-associated pathology, increase levels of hippocampal neurogenesis…

Figure 4.. FMD cycles slow the progression of AD-associated pathology, increase levels of hippocampal neurogenesis markers, and regulate microglia levels and activation in aged 3xTg mice
(A) Representative images showing Aβ immunoreactivity and AT8-positive neurons (recognizes abnormally phosphorylated tau) in subiculum or CA1 hippocampus regions of female control, FMD, and 4% PR 3xTg mice. (B) Quantification of subiculum Aβ load (%) for female 3xTg control (n = 21), FMD (n = 16), and 4% PR (n = 14) mice. (C) Quantification of CA1 Aβ load (%) for female 3xTg control (n = 20), FMD (n = 16), and 4% PR (n = 14) mice. (D) Quantification of AT8+ neurons in the subiculum and CA1 for female 3xTg control (n = 20), FMD (n = 14), and PR (n = 12) mice. (E) Representative images showing Aβ immunoreactivity and AT8+ neurons in subiculum or CA1 hippocampus regions of male 3xTg control, FMD, and 4% PR mice. (F) Quantification of subiculum Aβ load (%) for male 3xTg control (n = 15), FMD (n = 14), and 4%PR (n = 12) mice. (G) Quantification of CA1 Aβ load (%) for male 3xTg control (n = 15), FMD (n = 14), and 4%PR (n = 12) mice. (H) Quantification of AT8+ neurons in the subiculum and CA1 for male 3xTg control (n = 12), FMD (n = 13), and 4%PR (n = 11) mice. (I) Representative images showing BrdU+ DAB-immunohistochemistry in DG of hippocampus for 18.5-month-old female 3xTg control and FMD groups (left). Quantification of BrdU+ cells within the SGZ and inner third of the granule cell layer of the DG for 18.5-month-old female 3xTg control (n = 15) and FMD (n = 15) groups (right). (J) Representative images showing Sox2+, BrdU+, and co-stain hippocampal immunohistochemistry for 18.5-month-old female 3xTg control and FMD groups (left). Quantification of 18.5-month-old 3xTg female BrdU+Sox2+ (control [n = 9] and FMD [n = 8]) cells within the SGZ and inner third of the granule cell layer of the DG after ~15 months of FMD cycles (right). (K) Representative images showing CD11b-ir microglia in hippocampus sections of 18.5-month-old female C57B/6 WT, 3xTg control, and 3xTg FMD mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of C57B/6 WT, 3xTg control, and 3xTg FMD groups (bottom left; n = 5–7 animals per group). Percentage of different microglia activation stages (from 1 to 4) of C57B/6 WT, 3xTg control, and 3xTg FMD mice (bottom right; n = 5–7 animals per group). (L) Representative images showing BrdU+ DAB-immunohistochemistry in DG of hippocampus for 18.5-month-old male 3xTg control and FMD groups (left). Quantification of BrdU+ cells within the SGZ and inner third of the granule cell layer of the DG for 18.5-month-old male 3xTg control (n = 10) and FMD (n = 10) groups (right). (M) Representative images showing Sox2+, BrdU+, and co-stain hippocampal immunohistochemistry for 18.5-month-old male 3xTg control and FMD groups (left). Quantification of 18.5-month-old 3xTg male BrdU+Sox2+ (control [n = 11] and FMD [n = 9]) cells within the SGZ and inner third of the granule cell layer of the DG after ~15 months of FMD cycles (right). (N) Representative images showing CD11b-ir microglia in hippocampus sections of 18.5-month-old male C57B/6 WT, 3xTg control, and 3xTg FMD mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of C57B/6 WT, 3xTg control, and 3xTg FMD (bottom left; n = 5–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of C57B/6 WT, 3xTg control, and 3xTg FMD mice (bottom right; n = 5–8 animals per group). (O) Representative images showing CD68+ microglia in hippocampus sections of 18.5-month-old male 3xTg control and 3xTg FMD mice (top; n = 2–3 animals per group). Quantification of density of CD68+ cells in hippocampus CA1 and subiculum combined brain regions of 3xTg control and 3xTg FMD (bottom; n = 2–3 animals per group. Data are presented as mean ± SEM. (B–D and F–H) *p

Figure 5.. Short-term treatment with FMD cycles…

Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation,…

Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation, and reduces the expression of neuroinflammation genes, microglial activation, and reduced tau phosphorylation in 3xTg mice
(A) Experimental diet and behavior schedule for 3xTg males and females starting at 6.5 months of age through approximately 8.5 months of age for five FMD cycles. (B) RI for trial 2 of NOR task for 8.5-month-old C57B/6 WT males (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after four cycles of FMD and 7 days of refeeding (n = 5). (C) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg female control and FMD groups. (D) Quantification of subiculum Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (E) Quantification of CA1 Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (F) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg female control (n = 8) and FMD (n = 8) groups. (G) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg male control and FMD groups. (H) Quantification of subiculum Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (I) Quantification of CA1 Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (J) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg male control (n = 21) and FMD (n = 7) groups. (K) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old female C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom right). (L) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old male C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom right). (M) Representative images of confocal stack immune reactive for Iba1 microglia (red) in the prefrontal cortex for male and female 3xTg control diet and FMD cohorts. Scale bars, 100 μm. (N) Example 3D skeletonized microglial projections for 3xTg control diet and 3xTg FMD cohorts. (O and P) Quantification of Iba1 immuno-reactive soma area (O) and circularity (P) in male (M) and female (F) 3×Tg mice with control diet (open bars) and FMD (filled bars) from high-magnification images (n = 2/group). (Q) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old female 3xTg controls (n = 3) and 3xTg females after one 4-day cycle of FMD, with no refeeding (n = 6). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. (R) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old male 3xTg controls (n = 9) and 3xTg males after four cycles of FMD and 2 days of refeeding (n = 9). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. Data are presented as mean ± SEM. (B) *p

Figure 6.. Short-term cycles of FMD regulate…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice,…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice, while NOX2 deletion or inhibition improves cognitive behavior, mitigates pathology progression, and reduces microglia activation
(A) Diagram describing the dual positive and negative function of microglia. Microglia are involved in neuronal development and repair, but their production of O2− into ONOO− can promote toxicity and lead to neurodegeneration. (B) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female 3xTg controls (n = 4) and 3xTg females after one 4-day cycle of FMD with no refeeding (n = 10). (C) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female C57B/6 WT (n = 5) and 8.5-month-old 3xTg female control (n = 6) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (D) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old male C57B/6 WT (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (E) Quantification of Nox2 levels in cortex extract of control (n = 5) and FMD (n = 4) ~7-7.5-month-old female E4FAD mice after 4 months of biweekly FMD cycles (measured as Nox2/Vinculin protein expression levels). (F) Western blot image used for quantifying Nox2 levels in whole-cortex extract of control (n = 5) and FMD (n = 4) ~7- to 7.5-month-old female E4FAD mice. Vinculin was loading control (bottom). (G) 3xTg/Nox2-KO mice generation and experimental design. The experimental design of the tests conducted on 3xTg/Nox2-KO and control mice is depicted, as well as a schematic representation of the breeding strategy used to develop 3xTg/Nox2-KO mice and corresponding WT (mixed background 129/B6/B6). Some mice were euthanized for pathology at 13–14 months, while those used for fear-conditioning (FC) tests were aged until 15–18 months. (H) Representative images showing CD11b-ir microglia in hippocampus sections of 13- to 14-month-old male WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO (bottom left; n = 4–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (bottom right; n = 4–8 animals per group). (I) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice were tested with the Y-maze apparatus (12.5 months of age). SAB scores obtained through Y-maze task are shown (n = 7–23 per group). (J) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) FC tests. Freezing times (%) 24-h post shock for FC are shown (n = 6–22 per group) (left). WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) underwent FC tests. Freezing times (%) 48-h post shock for FC are shown (n = 6–22 per group) (right). (K) Representative images showing AT8 antibody (recognizes abnormally phosphorylated tau) immunoreactivity in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (left). Quantification of total AT8-immuno-reactive cells in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (n = 10–13/group) (right). (L) Experimental design of apocynin treatment. Eight-month-old WT (129/B6 background) and 3xTg mice were treated with apocynin-dissolved drinkable water or apocynin-free water for 6 months. During the final 4 weeks of treatment, the animals were tested using the Y-maze apparatus, and NOR and Rotarod assays. After completion of the behavioral tasks, the mice were euthanized and their brains analyzed. (M) Comparison of SAB scores between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice using the Y-maze apparatus (n = 10–13 animals per group). (N) Comparison of RI values between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin- treated mice during NOR test (n = 10–13 animals per group). (O) Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice (n = 5–10 animals/group). Data are presented as mean ± SEM. (H and O) *p

Figure 7.. Safety and feasibility of FMD…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI or mild AD
(A) A phase I/II randomized and placebo-controlled (single-blind) clinical study for 40 patients with aMCI or mild AD was designed for 12 monthly FMD cycles; 28/40 enrolled patients were randomly assigned to a placebo (control) diet arm (n = 16) or an FMD (ProLonAD) arm (n = 12). In the placebo arm, a gray square indicates that, for 5 days a month, patients are assigned a diet in which lunch or dinner is replaced with a meal based on pasta or rice with vegetables, without added supplements. In the FMD arm, a black square indicates a 5-day FMD cycle, followed by a striped box that indicates 25 days of a normal diet. White box indicates an in-between period between FMD cycles. Primary and secondary endpoints collected at each visit (per FMD cycles completed) is described in lower right-hand corner. (B) Graphical representation of the effects of FMD on the E4FAD and the 3xTg mouse models of AD, through which it mediates AD-associated pathology, neuroinflammation, NSCs, and Nox2 levels.
All figures (7)
Similar articles
References
    1. ‘t Hart BA, Copray S, and Philippens I (2014). Apocynin, a low molecular oral treatment for neurodegenerative disease. BioMed Res. Int, 298020. - PMC - PubMed
    1. Abels ER, Maas SLN, Nieland L, Wei Z, Cheah PS, Tai E, Kolsteeg CJ, Dusoswa SA, Ting DT, Hickman S, et al. (2019). Glioblastoma-associated microglia reprogramming is mediated by functional transfer of extracellular miR-21. Cell Rep. 28, 3105–3119.e7. - PMC - PubMed
    1. Alves S, Churlaud G, Audrain M, Michaelsen-Preusse K, Fol R, Souchet B, Braudeau J, Korte M, Klatzmann D, and Cartier N (2017). Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain 140, 826–842. - PubMed
    1. Anders S, Pyl PT, and Huber W (2015). HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169. - PMC - PubMed
    1. Anderson AJ, Cummings BJ, and Cotman CW (1994). Increased immunoreactivity for Jun- and Fos-related proteins in Alzheimer’s disease: association with pathology. Exp. Neurol 125, 286–295. - PubMed
Show all 135 references
Publication types
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 4.. FMD cycles slow the progression…
Figure 4.. FMD cycles slow the progression of AD-associated pathology, increase levels of hippocampal neurogenesis markers, and regulate microglia levels and activation in aged 3xTg mice
(A) Representative images showing Aβ immunoreactivity and AT8-positive neurons (recognizes abnormally phosphorylated tau) in subiculum or CA1 hippocampus regions of female control, FMD, and 4% PR 3xTg mice. (B) Quantification of subiculum Aβ load (%) for female 3xTg control (n = 21), FMD (n = 16), and 4% PR (n = 14) mice. (C) Quantification of CA1 Aβ load (%) for female 3xTg control (n = 20), FMD (n = 16), and 4% PR (n = 14) mice. (D) Quantification of AT8+ neurons in the subiculum and CA1 for female 3xTg control (n = 20), FMD (n = 14), and PR (n = 12) mice. (E) Representative images showing Aβ immunoreactivity and AT8+ neurons in subiculum or CA1 hippocampus regions of male 3xTg control, FMD, and 4% PR mice. (F) Quantification of subiculum Aβ load (%) for male 3xTg control (n = 15), FMD (n = 14), and 4%PR (n = 12) mice. (G) Quantification of CA1 Aβ load (%) for male 3xTg control (n = 15), FMD (n = 14), and 4%PR (n = 12) mice. (H) Quantification of AT8+ neurons in the subiculum and CA1 for male 3xTg control (n = 12), FMD (n = 13), and 4%PR (n = 11) mice. (I) Representative images showing BrdU+ DAB-immunohistochemistry in DG of hippocampus for 18.5-month-old female 3xTg control and FMD groups (left). Quantification of BrdU+ cells within the SGZ and inner third of the granule cell layer of the DG for 18.5-month-old female 3xTg control (n = 15) and FMD (n = 15) groups (right). (J) Representative images showing Sox2+, BrdU+, and co-stain hippocampal immunohistochemistry for 18.5-month-old female 3xTg control and FMD groups (left). Quantification of 18.5-month-old 3xTg female BrdU+Sox2+ (control [n = 9] and FMD [n = 8]) cells within the SGZ and inner third of the granule cell layer of the DG after ~15 months of FMD cycles (right). (K) Representative images showing CD11b-ir microglia in hippocampus sections of 18.5-month-old female C57B/6 WT, 3xTg control, and 3xTg FMD mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of C57B/6 WT, 3xTg control, and 3xTg FMD groups (bottom left; n = 5–7 animals per group). Percentage of different microglia activation stages (from 1 to 4) of C57B/6 WT, 3xTg control, and 3xTg FMD mice (bottom right; n = 5–7 animals per group). (L) Representative images showing BrdU+ DAB-immunohistochemistry in DG of hippocampus for 18.5-month-old male 3xTg control and FMD groups (left). Quantification of BrdU+ cells within the SGZ and inner third of the granule cell layer of the DG for 18.5-month-old male 3xTg control (n = 10) and FMD (n = 10) groups (right). (M) Representative images showing Sox2+, BrdU+, and co-stain hippocampal immunohistochemistry for 18.5-month-old male 3xTg control and FMD groups (left). Quantification of 18.5-month-old 3xTg male BrdU+Sox2+ (control [n = 11] and FMD [n = 9]) cells within the SGZ and inner third of the granule cell layer of the DG after ~15 months of FMD cycles (right). (N) Representative images showing CD11b-ir microglia in hippocampus sections of 18.5-month-old male C57B/6 WT, 3xTg control, and 3xTg FMD mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of C57B/6 WT, 3xTg control, and 3xTg FMD (bottom left; n = 5–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of C57B/6 WT, 3xTg control, and 3xTg FMD mice (bottom right; n = 5–8 animals per group). (O) Representative images showing CD68+ microglia in hippocampus sections of 18.5-month-old male 3xTg control and 3xTg FMD mice (top; n = 2–3 animals per group). Quantification of density of CD68+ cells in hippocampus CA1 and subiculum combined brain regions of 3xTg control and 3xTg FMD (bottom; n = 2–3 animals per group. Data are presented as mean ± SEM. (B–D and F–H) *p

Figure 5.. Short-term treatment with FMD cycles…

Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation,…

Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation, and reduces the expression of neuroinflammation genes, microglial activation, and reduced tau phosphorylation in 3xTg mice
(A) Experimental diet and behavior schedule for 3xTg males and females starting at 6.5 months of age through approximately 8.5 months of age for five FMD cycles. (B) RI for trial 2 of NOR task for 8.5-month-old C57B/6 WT males (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after four cycles of FMD and 7 days of refeeding (n = 5). (C) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg female control and FMD groups. (D) Quantification of subiculum Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (E) Quantification of CA1 Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (F) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg female control (n = 8) and FMD (n = 8) groups. (G) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg male control and FMD groups. (H) Quantification of subiculum Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (I) Quantification of CA1 Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (J) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg male control (n = 21) and FMD (n = 7) groups. (K) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old female C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom right). (L) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old male C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom right). (M) Representative images of confocal stack immune reactive for Iba1 microglia (red) in the prefrontal cortex for male and female 3xTg control diet and FMD cohorts. Scale bars, 100 μm. (N) Example 3D skeletonized microglial projections for 3xTg control diet and 3xTg FMD cohorts. (O and P) Quantification of Iba1 immuno-reactive soma area (O) and circularity (P) in male (M) and female (F) 3×Tg mice with control diet (open bars) and FMD (filled bars) from high-magnification images (n = 2/group). (Q) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old female 3xTg controls (n = 3) and 3xTg females after one 4-day cycle of FMD, with no refeeding (n = 6). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. (R) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old male 3xTg controls (n = 9) and 3xTg males after four cycles of FMD and 2 days of refeeding (n = 9). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. Data are presented as mean ± SEM. (B) *p

Figure 6.. Short-term cycles of FMD regulate…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice,…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice, while NOX2 deletion or inhibition improves cognitive behavior, mitigates pathology progression, and reduces microglia activation
(A) Diagram describing the dual positive and negative function of microglia. Microglia are involved in neuronal development and repair, but their production of O2− into ONOO− can promote toxicity and lead to neurodegeneration. (B) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female 3xTg controls (n = 4) and 3xTg females after one 4-day cycle of FMD with no refeeding (n = 10). (C) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female C57B/6 WT (n = 5) and 8.5-month-old 3xTg female control (n = 6) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (D) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old male C57B/6 WT (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (E) Quantification of Nox2 levels in cortex extract of control (n = 5) and FMD (n = 4) ~7-7.5-month-old female E4FAD mice after 4 months of biweekly FMD cycles (measured as Nox2/Vinculin protein expression levels). (F) Western blot image used for quantifying Nox2 levels in whole-cortex extract of control (n = 5) and FMD (n = 4) ~7- to 7.5-month-old female E4FAD mice. Vinculin was loading control (bottom). (G) 3xTg/Nox2-KO mice generation and experimental design. The experimental design of the tests conducted on 3xTg/Nox2-KO and control mice is depicted, as well as a schematic representation of the breeding strategy used to develop 3xTg/Nox2-KO mice and corresponding WT (mixed background 129/B6/B6). Some mice were euthanized for pathology at 13–14 months, while those used for fear-conditioning (FC) tests were aged until 15–18 months. (H) Representative images showing CD11b-ir microglia in hippocampus sections of 13- to 14-month-old male WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO (bottom left; n = 4–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (bottom right; n = 4–8 animals per group). (I) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice were tested with the Y-maze apparatus (12.5 months of age). SAB scores obtained through Y-maze task are shown (n = 7–23 per group). (J) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) FC tests. Freezing times (%) 24-h post shock for FC are shown (n = 6–22 per group) (left). WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) underwent FC tests. Freezing times (%) 48-h post shock for FC are shown (n = 6–22 per group) (right). (K) Representative images showing AT8 antibody (recognizes abnormally phosphorylated tau) immunoreactivity in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (left). Quantification of total AT8-immuno-reactive cells in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (n = 10–13/group) (right). (L) Experimental design of apocynin treatment. Eight-month-old WT (129/B6 background) and 3xTg mice were treated with apocynin-dissolved drinkable water or apocynin-free water for 6 months. During the final 4 weeks of treatment, the animals were tested using the Y-maze apparatus, and NOR and Rotarod assays. After completion of the behavioral tasks, the mice were euthanized and their brains analyzed. (M) Comparison of SAB scores between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice using the Y-maze apparatus (n = 10–13 animals per group). (N) Comparison of RI values between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin- treated mice during NOR test (n = 10–13 animals per group). (O) Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice (n = 5–10 animals/group). Data are presented as mean ± SEM. (H and O) *p

Figure 7.. Safety and feasibility of FMD…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI or mild AD
(A) A phase I/II randomized and placebo-controlled (single-blind) clinical study for 40 patients with aMCI or mild AD was designed for 12 monthly FMD cycles; 28/40 enrolled patients were randomly assigned to a placebo (control) diet arm (n = 16) or an FMD (ProLonAD) arm (n = 12). In the placebo arm, a gray square indicates that, for 5 days a month, patients are assigned a diet in which lunch or dinner is replaced with a meal based on pasta or rice with vegetables, without added supplements. In the FMD arm, a black square indicates a 5-day FMD cycle, followed by a striped box that indicates 25 days of a normal diet. White box indicates an in-between period between FMD cycles. Primary and secondary endpoints collected at each visit (per FMD cycles completed) is described in lower right-hand corner. (B) Graphical representation of the effects of FMD on the E4FAD and the 3xTg mouse models of AD, through which it mediates AD-associated pathology, neuroinflammation, NSCs, and Nox2 levels.
All figures (7)
Similar articles
References
    1. ‘t Hart BA, Copray S, and Philippens I (2014). Apocynin, a low molecular oral treatment for neurodegenerative disease. BioMed Res. Int, 298020. - PMC - PubMed
    1. Abels ER, Maas SLN, Nieland L, Wei Z, Cheah PS, Tai E, Kolsteeg CJ, Dusoswa SA, Ting DT, Hickman S, et al. (2019). Glioblastoma-associated microglia reprogramming is mediated by functional transfer of extracellular miR-21. Cell Rep. 28, 3105–3119.e7. - PMC - PubMed
    1. Alves S, Churlaud G, Audrain M, Michaelsen-Preusse K, Fol R, Souchet B, Braudeau J, Korte M, Klatzmann D, and Cartier N (2017). Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain 140, 826–842. - PubMed
    1. Anders S, Pyl PT, and Huber W (2015). HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169. - PMC - PubMed
    1. Anderson AJ, Cummings BJ, and Cotman CW (1994). Increased immunoreactivity for Jun- and Fos-related proteins in Alzheimer’s disease: association with pathology. Exp. Neurol 125, 286–295. - PubMed
Show all 135 references
Publication types
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 5.. Short-term treatment with FMD cycles…
Figure 5.. Short-term treatment with FMD cycles improves memory, mitigates pathology progression, reduces microglia activation, and reduces the expression of neuroinflammation genes, microglial activation, and reduced tau phosphorylation in 3xTg mice
(A) Experimental diet and behavior schedule for 3xTg males and females starting at 6.5 months of age through approximately 8.5 months of age for five FMD cycles. (B) RI for trial 2 of NOR task for 8.5-month-old C57B/6 WT males (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after four cycles of FMD and 7 days of refeeding (n = 5). (C) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg female control and FMD groups. (D) Quantification of subiculum Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (E) Quantification of CA1 Aβ load (%) for 3xTg female control (n = 7) and FMD (n = 7) groups. (F) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg female control (n = 8) and FMD (n = 8) groups. (G) Representative images showing subiculum and CA1 Aβ immunoreactivity and AT8+ neurons in hippocampus for 8.5-month-old 3xTg male control and FMD groups. (H) Quantification of subiculum Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (I) Quantification of CA1 Aβ load (%) for 3xTg male control (n = 20) and FMD (n = 6) groups. (J) Quantification of AT8+ neurons in the subiculum and CA1 for 3xTg male control (n = 21) and FMD (n = 7) groups. (K) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old female C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT females (n = 8) and 3xTg female control (n = 7) and FMD (n = 7) groups (bottom right). (L) Representative images showing Iba1-stained microglia in hippocampus sections of 8.5-month-old male C57B/6 WT and 3xTg control and FMD groups (top). Quantification of density of Iba1+ microglia in the CA1 and subiculum hippocampus regions of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom left). Percentage of different microglia activation stages (from 1, resting, to 4, most activated) of C57B/6 WT males (n = 8) and 3xTg male control (n = 16) and FMD (n = 5) groups (bottom right). (M) Representative images of confocal stack immune reactive for Iba1 microglia (red) in the prefrontal cortex for male and female 3xTg control diet and FMD cohorts. Scale bars, 100 μm. (N) Example 3D skeletonized microglial projections for 3xTg control diet and 3xTg FMD cohorts. (O and P) Quantification of Iba1 immuno-reactive soma area (O) and circularity (P) in male (M) and female (F) 3×Tg mice with control diet (open bars) and FMD (filled bars) from high-magnification images (n = 2/group). (Q) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old female 3xTg controls (n = 3) and 3xTg females after one 4-day cycle of FMD, with no refeeding (n = 6). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. (R) Experimental timeline and gene expression (top left and right) in cortex samples of 8.5-month-old male 3xTg controls (n = 9) and 3xTg males after four cycles of FMD and 2 days of refeeding (n = 9). Yellow-, green-, blue-, rose-, gray-, and lavender-highlighted gene names in heatmap correspond with genes associated with neuroinflammation, microglial activation, neuroinflammation and microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis, respectively. Relative fold change (Log2-transformed fold-change values, centered at 0) of gene expression in FMD group versus control diet group (bottom left) for genes associated with neuroinflammation, microglial activation, anti-neuroinflammation, tau phosphorylation, and general association with AD pathogenesis. Values in histogram were calculated from fold change in Table S1. “0” value is equivalent to no change between FMD group and control diet group. Data are presented as mean ± SEM. (B) *p

Figure 6.. Short-term cycles of FMD regulate…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice,…

Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice, while NOX2 deletion or inhibition improves cognitive behavior, mitigates pathology progression, and reduces microglia activation
(A) Diagram describing the dual positive and negative function of microglia. Microglia are involved in neuronal development and repair, but their production of O2− into ONOO− can promote toxicity and lead to neurodegeneration. (B) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female 3xTg controls (n = 4) and 3xTg females after one 4-day cycle of FMD with no refeeding (n = 10). (C) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female C57B/6 WT (n = 5) and 8.5-month-old 3xTg female control (n = 6) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (D) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old male C57B/6 WT (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (E) Quantification of Nox2 levels in cortex extract of control (n = 5) and FMD (n = 4) ~7-7.5-month-old female E4FAD mice after 4 months of biweekly FMD cycles (measured as Nox2/Vinculin protein expression levels). (F) Western blot image used for quantifying Nox2 levels in whole-cortex extract of control (n = 5) and FMD (n = 4) ~7- to 7.5-month-old female E4FAD mice. Vinculin was loading control (bottom). (G) 3xTg/Nox2-KO mice generation and experimental design. The experimental design of the tests conducted on 3xTg/Nox2-KO and control mice is depicted, as well as a schematic representation of the breeding strategy used to develop 3xTg/Nox2-KO mice and corresponding WT (mixed background 129/B6/B6). Some mice were euthanized for pathology at 13–14 months, while those used for fear-conditioning (FC) tests were aged until 15–18 months. (H) Representative images showing CD11b-ir microglia in hippocampus sections of 13- to 14-month-old male WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO (bottom left; n = 4–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (bottom right; n = 4–8 animals per group). (I) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice were tested with the Y-maze apparatus (12.5 months of age). SAB scores obtained through Y-maze task are shown (n = 7–23 per group). (J) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) FC tests. Freezing times (%) 24-h post shock for FC are shown (n = 6–22 per group) (left). WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) underwent FC tests. Freezing times (%) 48-h post shock for FC are shown (n = 6–22 per group) (right). (K) Representative images showing AT8 antibody (recognizes abnormally phosphorylated tau) immunoreactivity in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (left). Quantification of total AT8-immuno-reactive cells in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (n = 10–13/group) (right). (L) Experimental design of apocynin treatment. Eight-month-old WT (129/B6 background) and 3xTg mice were treated with apocynin-dissolved drinkable water or apocynin-free water for 6 months. During the final 4 weeks of treatment, the animals were tested using the Y-maze apparatus, and NOR and Rotarod assays. After completion of the behavioral tasks, the mice were euthanized and their brains analyzed. (M) Comparison of SAB scores between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice using the Y-maze apparatus (n = 10–13 animals per group). (N) Comparison of RI values between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin- treated mice during NOR test (n = 10–13 animals per group). (O) Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice (n = 5–10 animals/group). Data are presented as mean ± SEM. (H and O) *p

Figure 7.. Safety and feasibility of FMD…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI or mild AD
(A) A phase I/II randomized and placebo-controlled (single-blind) clinical study for 40 patients with aMCI or mild AD was designed for 12 monthly FMD cycles; 28/40 enrolled patients were randomly assigned to a placebo (control) diet arm (n = 16) or an FMD (ProLonAD) arm (n = 12). In the placebo arm, a gray square indicates that, for 5 days a month, patients are assigned a diet in which lunch or dinner is replaced with a meal based on pasta or rice with vegetables, without added supplements. In the FMD arm, a black square indicates a 5-day FMD cycle, followed by a striped box that indicates 25 days of a normal diet. White box indicates an in-between period between FMD cycles. Primary and secondary endpoints collected at each visit (per FMD cycles completed) is described in lower right-hand corner. (B) Graphical representation of the effects of FMD on the E4FAD and the 3xTg mouse models of AD, through which it mediates AD-associated pathology, neuroinflammation, NSCs, and Nox2 levels.
All figures (7)
Similar articles
References
    1. ‘t Hart BA, Copray S, and Philippens I (2014). Apocynin, a low molecular oral treatment for neurodegenerative disease. BioMed Res. Int, 298020. - PMC - PubMed
    1. Abels ER, Maas SLN, Nieland L, Wei Z, Cheah PS, Tai E, Kolsteeg CJ, Dusoswa SA, Ting DT, Hickman S, et al. (2019). Glioblastoma-associated microglia reprogramming is mediated by functional transfer of extracellular miR-21. Cell Rep. 28, 3105–3119.e7. - PMC - PubMed
    1. Alves S, Churlaud G, Audrain M, Michaelsen-Preusse K, Fol R, Souchet B, Braudeau J, Korte M, Klatzmann D, and Cartier N (2017). Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain 140, 826–842. - PubMed
    1. Anders S, Pyl PT, and Huber W (2015). HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169. - PMC - PubMed
    1. Anderson AJ, Cummings BJ, and Cotman CW (1994). Increased immunoreactivity for Jun- and Fos-related proteins in Alzheimer’s disease: association with pathology. Exp. Neurol 125, 286–295. - PubMed
Show all 135 references
Publication types
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6.. Short-term cycles of FMD regulate…
Figure 6.. Short-term cycles of FMD regulate Nox2 cortex levels in 3xTg and E4FAD mice, while NOX2 deletion or inhibition improves cognitive behavior, mitigates pathology progression, and reduces microglia activation
(A) Diagram describing the dual positive and negative function of microglia. Microglia are involved in neuronal development and repair, but their production of O2− into ONOO− can promote toxicity and lead to neurodegeneration. (B) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female 3xTg controls (n = 4) and 3xTg females after one 4-day cycle of FMD with no refeeding (n = 10). (C) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old female C57B/6 WT (n = 5) and 8.5-month-old 3xTg female control (n = 6) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (D) Quantification of Nox2 (ng/mL) in cortex extract of 8.5-month-old male C57B/6 WT (n = 8) and 8.5-month-old 3xTg male control (n = 10) and FMD after five cycles of FMD and no refeeding after last cycle (n = 4) groups. (E) Quantification of Nox2 levels in cortex extract of control (n = 5) and FMD (n = 4) ~7-7.5-month-old female E4FAD mice after 4 months of biweekly FMD cycles (measured as Nox2/Vinculin protein expression levels). (F) Western blot image used for quantifying Nox2 levels in whole-cortex extract of control (n = 5) and FMD (n = 4) ~7- to 7.5-month-old female E4FAD mice. Vinculin was loading control (bottom). (G) 3xTg/Nox2-KO mice generation and experimental design. The experimental design of the tests conducted on 3xTg/Nox2-KO and control mice is depicted, as well as a schematic representation of the breeding strategy used to develop 3xTg/Nox2-KO mice and corresponding WT (mixed background 129/B6/B6). Some mice were euthanized for pathology at 13–14 months, while those used for fear-conditioning (FC) tests were aged until 15–18 months. (H) Representative images showing CD11b-ir microglia in hippocampus sections of 13- to 14-month-old male WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (top). Quantification of density of CD11b-ir cells in hippocampus CA1 and subiculum combined brain regions of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO (bottom left; n = 4–8 animals per group). Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO mice (bottom right; n = 4–8 animals per group). (I) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice were tested with the Y-maze apparatus (12.5 months of age). SAB scores obtained through Y-maze task are shown (n = 7–23 per group). (J) WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) FC tests. Freezing times (%) 24-h post shock for FC are shown (n = 6–22 per group) (left). WT (129/B6 background), 3xTg, and 3xTg/Nox2-KO male mice (15–18 months of age) underwent FC tests. Freezing times (%) 48-h post shock for FC are shown (n = 6–22 per group) (right). (K) Representative images showing AT8 antibody (recognizes abnormally phosphorylated tau) immunoreactivity in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (left). Quantification of total AT8-immuno-reactive cells in hippocampus of 13- to 14-month-old male 3xTg and 3xTg/Nox2-KO mice (n = 10–13/group) (right). (L) Experimental design of apocynin treatment. Eight-month-old WT (129/B6 background) and 3xTg mice were treated with apocynin-dissolved drinkable water or apocynin-free water for 6 months. During the final 4 weeks of treatment, the animals were tested using the Y-maze apparatus, and NOR and Rotarod assays. After completion of the behavioral tasks, the mice were euthanized and their brains analyzed. (M) Comparison of SAB scores between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice using the Y-maze apparatus (n = 10–13 animals per group). (N) Comparison of RI values between WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin- treated mice during NOR test (n = 10–13 animals per group). (O) Percentage of different microglia activation stages (from 1 to 4) of WT (129/B6 background) vehicle, 3xTg vehicle, and 3xTg apocynin-treated mice (n = 5–10 animals/group). Data are presented as mean ± SEM. (H and O) *p

Figure 7.. Safety and feasibility of FMD…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI…

Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI or mild AD
(A) A phase I/II randomized and placebo-controlled (single-blind) clinical study for 40 patients with aMCI or mild AD was designed for 12 monthly FMD cycles; 28/40 enrolled patients were randomly assigned to a placebo (control) diet arm (n = 16) or an FMD (ProLonAD) arm (n = 12). In the placebo arm, a gray square indicates that, for 5 days a month, patients are assigned a diet in which lunch or dinner is replaced with a meal based on pasta or rice with vegetables, without added supplements. In the FMD arm, a black square indicates a 5-day FMD cycle, followed by a striped box that indicates 25 days of a normal diet. White box indicates an in-between period between FMD cycles. Primary and secondary endpoints collected at each visit (per FMD cycles completed) is described in lower right-hand corner. (B) Graphical representation of the effects of FMD on the E4FAD and the 3xTg mouse models of AD, through which it mediates AD-associated pathology, neuroinflammation, NSCs, and Nox2 levels.
All figures (7)
Figure 7.. Safety and feasibility of FMD…
Figure 7.. Safety and feasibility of FMD cycles as a treatment for patients with aMCI or mild AD
(A) A phase I/II randomized and placebo-controlled (single-blind) clinical study for 40 patients with aMCI or mild AD was designed for 12 monthly FMD cycles; 28/40 enrolled patients were randomly assigned to a placebo (control) diet arm (n = 16) or an FMD (ProLonAD) arm (n = 12). In the placebo arm, a gray square indicates that, for 5 days a month, patients are assigned a diet in which lunch or dinner is replaced with a meal based on pasta or rice with vegetables, without added supplements. In the FMD arm, a black square indicates a 5-day FMD cycle, followed by a striped box that indicates 25 days of a normal diet. White box indicates an in-between period between FMD cycles. Primary and secondary endpoints collected at each visit (per FMD cycles completed) is described in lower right-hand corner. (B) Graphical representation of the effects of FMD on the E4FAD and the 3xTg mouse models of AD, through which it mediates AD-associated pathology, neuroinflammation, NSCs, and Nox2 levels.

References

    1. ‘t Hart BA, Copray S, and Philippens I (2014). Apocynin, a low molecular oral treatment for neurodegenerative disease. BioMed Res. Int, 298020.
    1. Abels ER, Maas SLN, Nieland L, Wei Z, Cheah PS, Tai E, Kolsteeg CJ, Dusoswa SA, Ting DT, Hickman S, et al. (2019). Glioblastoma-associated microglia reprogramming is mediated by functional transfer of extracellular miR-21. Cell Rep. 28, 3105–3119.e7.
    1. Alves S, Churlaud G, Audrain M, Michaelsen-Preusse K, Fol R, Souchet B, Braudeau J, Korte M, Klatzmann D, and Cartier N (2017). Interleukin-2 improves amyloid pathology, synaptic failure and memory in Alzheimer’s disease mice. Brain 140, 826–842.
    1. Anders S, Pyl PT, and Huber W (2015). HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169.
    1. Anderson AJ, Cummings BJ, and Cotman CW (1994). Increased immunoreactivity for Jun- and Fos-related proteins in Alzheimer’s disease: association with pathology. Exp. Neurol 125, 286–295.
    1. Andrade J, Quinn J, Becker RZ, and Shupnik MA (2013). AMP-activated protein kinase is a key intermediary in GnRH-stimulated LHbeta gene transcription. Mol. Endocrinol 27, 828–839.
    1. Andrews S (2010). FastQC: A Quality Control Tool for High Throughput Sequence Data. .
    1. Azevedo H, Khaled NA, Santos P, Bertonha FB, and Moreira-Filho CA (2018). Temporal analysis of hippocampal CA3 gene co-expression networks in a rat model of febrile seizures. Disease models & mechanisms 11, 1–12.
    1. Bachmanov AA, Reed DR, Beauchamp GK, and Tordoff MG (2002). Food intake, water intake, and drinking spout side preference of 28 mouse strains. Behav. Genet 32, 435–443.
    1. Baillie JK, Arner E, Daub C, De Hoon M, Itoh M, Kawaji H, Lassmann T, Carninci P, Forrest ARR, Hayashizaki Y, et al. (2017). Analysis of the human monocyte-derived macrophage transcriptome and response to lipopolysaccharide provides new insights into genetic aetiology of inflammatory bowel disease. PLoS Genet. 13, e1006641.
    1. Barnes CA (1988). Spatial learning and memory processes: the search for their neurobiological mechanisms in the rat. Trends Neurosci. 11, 163–169.
    1. Benboubker L, Dimopoulos MA, Dispenzieri A, Catalano J, Belch AR, Cavo M, Pinto A, Weisel K, Ludwig H, Bahlis N, et al. (2014). Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N. Engl. J. Med 371, 906–917.
    1. Berasi SP, Huard C, Li D, Shih HH, Sun Y, Zhong W, Paulsen JE, Brown EL, Gimeno RE, and Martinez RV (2006). Inhibition of gluconeogenesis through transcriptional activation of EGR1 and DUSP4 by AMP-activated kinase. J. Biol. Chem 281, 27167–27177.
    1. Biechele G, Franzmeier N, Blume T, Ewers M, Luque JM, Eckenweber F, Sacher C, Beyer L, Ruch-Rubinstein F, Lindner S, et al. (2020). Glial activation is moderated by sex in response to amyloidosis but not to tau pathology in mouse models of neurodegenerative diseases. J. Neuroinflammation 17, 374.
    1. Blagosklonny MV (2009). Validation of anti-aging drugs by treating age-related diseases. Aging 1, 281–288.
    1. Block ML (2008). NADPH oxidase as a therapeutic target in Alzheimer’s disease. BMC Neurosci. 9 (Suppl 2), S8.
    1. Bloom GS (2014). Amyloid-beta and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol. 71, 505–508.
    1. Bolger AM, Lohse M, and Usadel B (2014). Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120.
    1. Bonam SR, Ruff M, and Muller S (2019). HSPA8/HSC70 in immune disorders: a molecular rheostat that adjusts chaperone-mediated autophagy substrates. Cells 8, 849.
    1. Brandhorst S, Choi IY, Wei M, Cheng CW, Sedrakyan S, Navarrete G, Dubeau L, Yap LP, Park R, Vinciguerra M, et al. (2015). A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metab. 22, 86–99.
    1. Bruce-Keller AJ, Gupta S, Knight AG, Beckett TL, McMullen JM, Davis PR, Murphy MP, Van Eldik LJ, St Clair D, and Keller JN (2011). Cognitive impairment in humanized APP×PS1 mice is linked to Aβ (1-42) and NOX activation. Neurobiol. Dis 44, 317–326.
    1. Caccamo A, Magrì A, Medina DX, Wisely EV, López-Aranda MF, Silva AJ, and Oddo S (2013). mTOR regulates tau phosphorylation and degradation: implications for Alzheimer’s disease and other tauopathies. Aging Cell 12, 370–380.
    1. Cacciottolo M, Christensen A, Moser A, Liu J, Pike CJ, Smith C, LaDu MJ, Sullivan PM, Morgan TE, Dolzhenko E, et al. (2016). The APOE4 allele shows opposite sex bias in microbleeds and Alzheimer’s disease of humans and mice. Neurobiol. Aging 37, 47–57.
    1. Caffa I, Spagnolo V, Vernieri C, Valdemarin F, Becherini P, Wei M, Brandhorst S, Zucal C, Driehuis E, Ferrando L, et al. (2020). Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature 583, 620–624.
    1. Carlson M (2019). GO.db: a Set of Annotation Maps Describing the Entire Gene Ontology. R package version 3.8.2
    1. Carroll JC, Rosario ER, Villamagna A, and Pike CJ (2010). Continuous and cyclic progesterone differentially interact with estradiol in the regulation of Alzheimer like pathology in female 3x Transgenic-Alzheimer’s disease mice. Endocrinology 151, 2713–2722.
    1. Chen L, and Liu B (2017). Relationships between stress granules, oxidative stress, and neurodegenerative diseases. Oxid. Med. Cell. Longev, 1809592–1809610.
    1. Cheng CW, Villani V, Buono R, Wei M, Kumar S, Yilmaz OH, Cohen P, Sneddon JB, Perin L, and Longo VD (2017). Fasting-mimicking diet Promotes Ngn3-Driven β-Cell regeneration to reverse diabetes. Cell 168, 775–788.e12.
    1. Choi IY, Piccio L, Childress P, Bollman B, Ghosh A, Brandhorst S, Suarez J, Michalsen A, Cross AH, Morgan TE, et al. (2016). A diet mimicking fasting promotes regeneration and reduces autoimmunity and multiple sclerosis symptoms. Cell Rep. 15, 2136–2146.
    1. Chow N, Bell RD, Deane R, Streb JW, Chen J, Brooks A, Van Nostrand W, Miano JM, and Zlokovic BV (2007). Serum response factor and myocardin mediate arterial hypercontractility and cerebral blood flow dysregulation in Alzheimer’s phenotype. Proc. Natl. Acad. Sci. USA 104, 823–828.
    1. Chu J, Li JG, Ceballos-Diaz C, Golde T, and Praticò D (2013). The influence of 5-lipoxygenase on Alzheimer’s disease-related tau pathology: in vivo and in vitro evidence. Biol. Psychiatry 74, 321–328.
    1. Clarimón J, Bertranpetit J, Boada M, Tàrraga L, and Comas D (2003). HSP70-2 (HSPA1B) is associated with noncognitive symptoms in late-onset Alzheimer’s disease. J. Geriatr. Psychiatry Neurol 16, 146–150.
    1. Cline EN, Bicca MA, Viola KL, and Klein WL (2018). The amyloid-beta oligomer hypothesis: beginning of the third decade. J. Alzheimers Dis 64, S567–S610.
    1. Close AF, Dadheech N, Villela BS, Rouillard C, and Buteau J (2019). The orphan nuclear receptor Nor1/Nr4a3 is a negative regulator of β-cell mass. J. Biol. Chem 294, 4889–4897.
    1. Connelly PJ, Adams F, Tayar ZI, and Khan F (2019). Peripheral vascular responses to acetylcholine as a predictive tool for response to cholinesterase inhibitors in Alzheimer’s disease. BMC Neurol. 19, 88.
    1. Crews FT, and Vetreno RP (2016). Mechanisms of neuroimmune gene induction in alcoholism. Psychopharmacology 233, 1543–1557.
    1. Davis BM, Salinas-Navarro M, Cordeiro MF, Moons L, and De Groef L (2017). Characterizing microglia activation: a spatial statistics approach to maximize information extraction. Sci. Rep 7.
    1. Di Biase S, Shim HS, Kim KH, Vinciguerra M, Rappa F, Wei M, Brandhorst S, Cappello F, Mirzaei H, Lee C, and Longo VD (2017). Fasting regulates EGR1 and protects from glucose- and dexamethasone-dependent sensitization to chemotherapy. PLoS Biol. 15, e2001951.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, and Gingeras TR (2013). Star : ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21.
    1. Dong Y, Dekens D, De Deyn P, Naudé P, and Eisel U (2015). Targeting of tumor necrosis factor alpha receptors as a therapeutic strategy for neurodegenerative disorders. Antibodies 4, 369–408.
    1. Draznin B, Wang C, Adochio R, Leitner JW, and Cornier MA (2012). Effect of dietary macronutrient composition on AMPK and SIRT1 expression and activity in human skeletal muscle. Horm. Metab. Res 44, 650–655.
    1. Fang Y, Yao L, Li C, Wang J, Wang J, Chen S, Zhou XF, and Liao H (2016). The blockage of the Nogo/NgR signal pathway in microglia alleviates the formation of Aβ plaques and tau phosphorylation in APP/PS1 transgenic mice. J. Neuroinflammation 13, 56.
    1. Flood WD, Moyer RW, Tsykin A, Sutherland GR, and Koblar SA (2004). Nxf and Fbxo33: novel seizure-responsive genes in mice. Eur. J. Neurosci 20, 1819–1826.
    1. Gao W, Li F, Zhou Z, Xu X, Wu Y, Zhou S, Yin D, Sun D, Xiong J, Jiang R, and Zhang J (2017). IL-2/Anti-IL-2 complex attenuates inflammation and BBB disruption in mice subjected to traumatic brain injury. Front. Neurol 8, 281.
    1. Giri RK, Rajagopal V, Shahi S, Zlokovic BV, and Kalra VK (2005). Mechanism of amyloid peptide induced CCR5 expression in monocytes and its inhibition by siRNA for Egr-1. Am. J. Physiol. Cell Physiol 289, 264–276.
    1. Gong Y, Chang L, Viola KL, Lacor PN, Lambert MP, Finch CE, Krafft GA, and Klein WL (2003). Alzheimer’s disease-affected brain: presence of oligomeric A beta ligands (ADDLs) suggests a molecular basis for reversible memory loss. Proc. Natl. Acad. Sci. USA 100, 10417–10422.
    1. Guo Q, Fu W, Sopher BL, Miller MW, Ware CB, Martin GM, and Mattson MP (1999). Increased vulnerability of hippocampal neurons to excitotoxic necrosis in presenilin-1 mutant knock-in mice. Nat. Med 5, 101–106.
    1. Halagappa VKM, Guo Z, Pearson M, Matsuoka Y, Cutler RG, Laferla FM, and Mattson MP (2007). Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer’s disease. Neurobiol. Dis 26, 212–220.
    1. Hardie DG (2011). AMP-activated protein kinase - an energy sensor that regulates all aspects of cell function. Genes Dev. 25, 1895–1908.
    1. Harraz MM, Marden JJ, Zhou W, Zhang Y, Williams A, Sharov VS, Nelson K, Luo M, Paulson H, Schöneich C, and Engelhardt JF (2008). SOD1 mutations disrupt redox-sensitive Rac regulation of NADPH oxidase in a familial ALS model. J. Clin. Invest 118, 659–670.
    1. Herrmann L, Wiegmann C, Arsalan-Werner A, Hilbrich I, Jäger C, Flach K, Suttkus A, Lachmann I, Arendt T, and Holzer M (2015). Hook proteins: association with Alzheimer pathology and regulatory role of hook3 in amyloid beta generation. PLoS One 10, e0119423.
    1. Hovens I, Nyakas C, and Schoemaker R (2014). A novel method for evaluating microglial activation using ionized calcium-binding adaptor protein-1 staining: cell body to cell size ratio. Neuroimmunol. Neuroinflamm 1, 82. .
    1. Jekabsone A, Mander PK, Tickler A, Sharpe M, and Brown GC (2006). Fibrillar beta-amyloid peptide Abeta1-40 activates microglial proliferation via stimulating TNF-alpha release and H2O2 derived from NADPH oxidase: a cell culture study. J. Neuroinflammation 3, 24.
    1. Kee N, Sivalingam S, Boonstra R, and Wojtowicz JM (2002). The utility of Ki-67 and BrdU as proliferative markers of adult neurogenesis. J. Neurosci. Methods 115, 97–105.
    1. Khuu MA, Pagan CM, Nallamothu T, Hevner RF, Hodge RD, Ramirez JM, and Garcia AJ (2018). Intermittent hypoxia disrupts adult neurogenesis and synaptic plasticity in the dentate gyrus. J. Neurosci 39, 1320–1331.
    1. Kostylev MA, Kaufman AC, Nygaard HB, Patel P, Haas LT, Gunther EC, Vortmeyer A, and Strittmatter SM (2015). Prion-Protein-interacting amyloid-β oligomers of high molecular weight are tightly correlated with memory impairment in multiple Alzheimer mouse models. J. Biol. Chem 290, 17415–17438.
    1. Kreutzberg GW (1996). Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 19, 312–318.
    1. Kuhn DM, Francescutti-Verbeem DM, and Thomas DM (2006). Dopamine quinones activate microglia and induce a neurotoxic gene expression profile. Ann N Y Acad Sci 1074, 31–41.
    1. Lafourcade CA, Lin TV, Feliciano DM, Zhang L, Hsieh LS, and Bordey A (2013). Rheb activation in subventricular zone progenitors leads to heterotopia, ectopic neuronal differentiation, and rapamycin-sensitive olfactory micronodules and dendrite hypertrophy of newborn neurons. J. Neurosci 33, 2419–2431.
    1. Levine ME, Suarez JA, Brandhorst S, Balasubramanian P, Cheng CW, Madia F, Fontana L, Mirisola MG, Guevara-Aguirre J, Wan J, et al. (2014). Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 19, 407–417.
    1. Lewandowski CT, Maldonado Weng J, and LaDu MJ (2020). Alzheimer’s disease pathology in ApoE transgenic mouse models: the who, what, when, where, why, and how. Neurobiol. Dis 139.
    1. Liu H, Jiang J, and Zhao L (2019). Protein arginine methyltransferase-1 deficiency restrains depression-like behavior of mice by inhibiting inflammation and oxidative stress via Nrf-2. Biochem. Biophys. Res. Commun 518, 430–437.
    1. Liu IYC, Lyons WE, Mamounas LA, and Thompson RF (2004). Brain-derived neurotrophic factor plays a critical role in contextual fear conditioning. J. Neurosci 24, 7958–7963.
    1. Lo AC, Iscru E, Blum D, Tesseur I, Callaerts-Vegh Z, Buée L, De Strooper B, Balschun D, and Dhooge R (2013). Amyloid and tau neuropathology differentially affect prefrontal synaptic plasticity and cognitive performance in mouse models of alzheimers disease. J. Alzheimers Dis 37, 109–125.
    1. Longo VD, Viola KL, Klein WL, and Finch CE (2000). Reversible inactivation of superoxide-sensitive aconitase in Abeta1-42-treated neuronal cell lines. J. Neurochem 75, 1977–1985.
    1. Lopes S, Teplytska L, Vaz-Silva J, Dioli C, Trindade R, Morais M, Webhofer C, Maccarrone G, Almeida OFX, Turck CW, et al. (2016). Tau deletion prevents stress-induced dendritic atrophy in prefrontal cortex: role of synaptic mitochondria. Cereb. Cortex 27, 2580–2591.
    1. Lynch MA (2009). The multifaceted profile of activated microglia. Mol. Neurobiol 40, 139–156.
    1. Lyons MR, and West AE (2011). Mechanisms of specificity in neuronal activity-regulated gene transcription. Prog. Neurobiol 94, 259–295.
    1. Ma CY, Yao M.j., Zhai Q.w., Jiao J.w., Yuan X.b., and Poo M.m. (2014). SIRT1 suppresses self-renewal of adult hippocampal neural stem cells. Development 141, 4697–4709.
    1. Magno L, Lessard CB, Martins M, Lang V, Cruz P, Asi Y, Katan M, Bilsland J, Lashley T, Chakrabarty P, et al. (2018). Alzheimer’s disease Phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph. Alzheimer’s Res. Ther 11.
    1. Maldonado Weng J, Parikh I, Naqib A, York J, Green SJ, Estus S, and LaDu MJ (2019). Synergistic effects of APOE and sex on the gut microbiome of young EFAD transgenic mice. Mol. Neurodegener 14.
    1. Malik M, Parikh I, Vasquez JB, Smith C, Tai L, Bu G, LaDu MJ, Fardo DW, Rebeck GW, and Estus S (2015). Genetics ignite focus on microglial inflammation in Alzheimer’s disease. Mol. Neurodegener 10, 52.
    1. Marballi KK, and Gallitano AL (2018). Immediate early genes anchor a biological pathway of proteins required for memory formation, long-term depression and risk for schizophrenia. Front. Behav. Neurosci 12.
    1. Massaad CA, and Klann E (2011). Reactive oxygen species in the regulation of synaptic plasticity and memory. Antioxid. Redox Signal 14, 2013–2054.
    1. Mastrangelo MA, and Bowers WJ (2008). Detailed immunohistochemical characterization of temporal and spatial progression of Alzheimer’s disease-related pathologies in male triple-transgenic mice. BMC Neurosci. 9, 81.
    1. McCarthy DJ, Chen Y, and Smyth GK (2012). Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297.
    1. McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR Jr., Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, et al. (2011). The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 7, 263–269.
    1. Méndez-Samperio P (2010). Role of interleukin-12 family cytokines in the cellular response to mycobacterial disease. Int. J. Infect. Dis 14, e366–e371.
    1. Mengozzi M, Cervellini I, Villa P, Erbayraktar Z, Gökmen N, Yilmaz O, Erbayraktar S, Manohasandra M, Van Hummelen P, Vandenabeele P, et al. (2012). Erythropoietin-induced changes in brain gene expression reveal induction of synaptic plasticity genes in experimental stroke. Proc. Natl. Acad. Sci. USA 109, 9617–9622.
    1. Michán S, Li Y, Chou MMH, Parrella E, Ge H, Long JM, Allard JS, Lewis K, Miller M, Xu W, et al. (2010). SIRT1 is essential for normal cognitive function and synaptic plasticity. J. Neurosci 30, 9695–9707.
    1. Ming G.l., and Song H (2005). Adult neurogenesis in the mammalian central nervous system. Annu. Rev. Neurosci 28, 223–250.
    1. Mirzaei H, Di Biase S, and Longo VD (2016). Dietary interventions, cardiovascular aging, and disease: animal models and human studies. Circ. Res 118, 1612–1625.
    1. Mori F, Nishie M, Piao YS, Kito K, Kamitani T, Takahashi H, and Wakabayashi K (2004). Accumulation of NEDD8 in neuronal and glial inclusions of neurodegenerative disorders. Neuropathol. Appl. Neurobiol 31, 53–61.
    1. Mouton PR, Chachich ME, Quigley C, Spangler E, and Ingram DK (2009). Caloric restriction attenuates amyloid deposition in middle-aged dtg APP/PS1 mice. Neurosci. Lett 464, 184–187.
    1. Nomaru H, Sakumi K, Katogi A, Ohnishi YN, Kajitani K, Tsuchimoto D, Nestler EJ, and Nakabeppu Y (2015). Fosb gene products contribute to excitotoxic microglial activation by regulating the expression of complement C5a receptors in microglia. Glia 62, 1284–1298.
    1. Nomura K, Vilalta A, Allendorf DH, Hornik TC, and Brown GC (2017). Activated microglia desialylate and phagocytose cells via neuraminidase, galectin-3, and mer tyrosine kinase. J. Immunol 198, 4792–4801.
    1. Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-Bongaarts A, Ohno M, Disterhoft J, Van Eldik L, et al. (2006). Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J. Neurosci 26, 10129–10140.
    1. Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R, Metherate R, Mattson MP, Akbari Y, and LaFerla FM (2003). Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron 39, 409–421.
    1. Pandey RS, Graham L, Uyar A, Preuss C, Howell GR, and Carter GW (2019). Genetic perturbations of disease risk genes in mice capture transcriptomic signatures of late-onset Alzheimer’s disease. Mol. Neurodegener 14, 50.
    1. Papadopoulos C, Kirchner P, Bug M, Grum D, Koerver L, Schulze N, Poehler R, Dressler A, Fengler S, Arhzaouy K, et al. (2017). VCP/p97 cooperates with YOD1, UBXD1 and PLAA to drive clearance of ruptured lysosomes by autophagy. EMBO J. 36, 135–150.
    1. Park HJ, Lee KW, Oh S, Yan R, Zhang J, Beach TG, Adler CH, Voronkov M, Braithwaite SP, Stock JB, and Mouradian MM (2018). Protein phosphatase 2A and its methylation modulating enzymes LCMT-1 and PME-1 are dysregulated in tauopathies of progressive supranuclear palsy and Alzheimer’s disease. J. Neuropathol. Exp. Neurol 77, 139–148.
    1. Park J, Choi H, Kim B, Chae U, Lee DG, Lee SR, Lee S, Lee HS, and Lee DS (2016). Peroxiredoxin 5 (Prx5) decreases LPS-induced microglial activation through regulation of Ca2+/calcineurin-Drp1-dependent mitochondrial fission. Free Radic. Biol. Med 99, 392–404.
    1. Park L, Uekawa K, Garcia-Bonilla L, Koizumi K, Murphy M, Pistik R, Younkin L, Younkin S, Zhou P, Carlson G, et al. (2017). Brain perivascular macrophages initiate the neurovascular dysfunction of alzheimer Aβ peptides. Circ. Res 121, 258–269.
    1. Park L, Zhou P, Pitstick R, Capone C, Anrather J, Norris EH, Younkin L, Younkin S, Carlson G, McEwen BS, and Iadecola C (2008). Nox2-derived radicals contribute to neurovascular and behavioral dysfunction in mice overexpressing the amyloid precursor protein. Proc. Natl. Acad. Sci. U. S. A 105, 1347–1352.
    1. Park SY, Jung MY, Lee SJ, Kang KB, Gratchev A, Riabov V, Kzhyshkowska J, Kim IS, and Kim I (2009). Stabilin-1 mediates phosphatidylserine-dependent clearance of cell corpses in alternatively activated macrophages. J. Cell Sci 122, 3365–3373.
    1. Parrella E, Maxim T, Maialetti F, Zhang L, Wan J, Wei M, Cohen P, Fontana L, and Longo VD (2013). Protein restriction cycles reduce IGF-1 and phosphorylated tau, and improve behavioral performance in an alzheimer’s disease mouse model. Aging Cell 12, 257–268.
    1. Patel NV, Gordon MN, Connor KE, Good RA, Engelman RW, Mason J, Morgan DG, Morgan TE, and Finch CE (2005). Caloric restriction attenuates Aβ-deposition in alzheimer transgenic models. Neurobiol. Aging 26, 995–1000.
    1. Petersen RC (2004). Mild cognitive impairment as a diagnostic entry. J. Intern. Med 256, 183–194.
    1. Petitto JM, Jackson AJ, McNamara RK, Gendreau PL, and Huang Z (1999). Impaired learning and memory and altered hippocampal neurodevelopment resulting from interleukin-2 gene deletion. J. Neurosci. Res 56, 441–446.
    1. Plotkin SS, and Cashman NR (2020). Passive immunotherapies targeting Aβ and tau in Alzheimer’s disease. Neurobiol. Dis 144, 105010.
    1. Pollock JD, Williams DA, Gifford MA, Li LL, Du X, Fisherman J, Orkin SH, Doerschuk CM, and Dinauer MC (1995). Mouse model of X-linked chronic granulomatous disease, an inherited defect in phagocyte superoxide production. Nat. Genet 9, 202–209.
    1. Qin X, Wang Y, and Paudel HK (2016). Early growth response 1 (Egr-1) is a transcriptional activator of β-secretase 1 (BACE-1) in the brain. J. Biol. Chem 291, 22276–22287.
    1. Raj DDA, Moser J, Pol SMA, Os RP, Holtman IR, Brouwer N, Oeseburg H, Schaafsma W, Wesseling EM, Dunnen W, et al. (2015). Enhanced microglial pro-inflammatory response to lipopolysaccharide correlates with brain infiltration and blood–brain barrier dysregulation in a mouse model of telomere shortening. Aging Cell 14, 1003–1013.
    1. Rangan P, Choi I, Wei M, Navarrete G, Guen E, Brandhorst S, Enyati N, Pasia G, Maesincee D, Ocon V, et al. (2019). Fasting-mimicking diet modulates microbiota and promotes intestinal regeneration to reduce inflammatory bowel disease pathology. Cell Rep. 26, 2704–2719.e6.
    1. Reimand J, Kull M, Peterson H, Hansen J, and Vilo J (2007). g: Profiler–a web-based toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res. 35, W193–W200.
    1. Robinson MD, McCarthy DJ, and Smyth GK (2010). edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140.
    1. Rodríguez JJ, Jones VC, Tabuchi M, Allan SM, Knight EM, Laferla FM, Oddo S, and Verkhratsky A (2008). Impaired adult neurogenesis in the dentate gyrus of a triple transgenic mouse model of alzheimer’s disease. PLoS One 3, e2935.
    1. Rosi S (2011). Neuroinflammation and the plasticity-related immediate-early gene Arc. Brain Behav. Immun 25, 39–49.
    1. Sala Frigerio C, Wolfs L, Fattorelli N, Thrupp N, Voytyuk I, Schmidt I, Mancuso R, Chen WT, Woodbury ME, Srivastava G, et al. (2019). The major risk factors for alzheimer’s disease: age, sex, and genes modulate the microglia response to Aβ plaques. Cell Rep. 27, 1293–1306.e6.
    1. Schlachetzki JCM, and Hüll M (2009). Microglial activation in alzheimer’s disease. Curr. Alzheimer Res 6, 554–563.
    1. Schneider LS, Dagerman KS, Higgins JPT, and McShane R (2011). Lack of evidence for the efficacy of memantine in mild alzheimer disease. Arch. Neurol 68, 991–998.
    1. Simonyi A, Serfozo P, Lehmidi TM, Cui J, Gu Z, Lubahn DB, Sun AY, and Sun GY (2012). The neuroprotective effects of apocynin. Front. Biosci 4, 2183–2193.
    1. Stefanska J, and Pawliczak R (2008). Apocynin: Molecular Aptitudes (Mediators of inflammation), p. 106507.
    1. Stephen T-L, Cacciottolo M, Balu D, Morgan TE, LaDu MJ, Finch CE, and Pike CJ (2019). APOE genotype and sex affect microglial interactions with plaques in Alzheimer’s disease mice. Acta neuropathologica communications 7, 82.
    1. Stephen T-L, Higgs NF, Sheehan DF, Al Awabdh S, López-Doménech G, Arancibia-Carcamo IL, and Kittler JT (2015). Miro1 regulates activity-driven positioning of mitochondria within astrocytic processes apposed to synapses to regulate intracellular calcium signaling. J. Neurosci 35, 15996–16011.
    1. Stephens KE, Zhou W, Ji Z, Chen Z, He S, Ji H, Guan Y, and Taverna SD (2019). Sex differences in gene regulation in the dorsal root ganglion after nerve injury. BMC Genom. 20, 147.
    1. Sterniczuk R, Antle MC, Laferla FM, and Dyck RH (2010). Characterization of the 3xTg mouse model of alzheimers disease: part 2. behavioral and cognitive changes. Brain Res. 1348, 149–155.
    1. Sun HN, Kim SU, Huang SM, Kim JM, Park YH, Kim SH, Yang HY, Chung KJ, Lee TH, Choi HS, et al. (2010). Microglial peroxiredoxin V acts as an inducible anti-inflammatory antioxidant through cooperation with redox signaling cascades. J. Neurochem 114, 39–50.
    1. Tamboli IY, Barth E, Christian L, Siepmann M, Kumar S, Singh S, Tolksdorf K, Heneka MT, Lütjohann D, Wunderlich P, and Walter J (2010). Statins promote the degradation of extracellular amyloid β-peptide by microglia via stimulation of exosome-associated IDE secretion. J. Biol. Chem 285, 37405–37414.
    1. Vázquez-Higuera JL, Mateo I, Sánchez-Juan P, Rodríguez-Rodríguez E, Pozueta A, Calero M, Dobato JL, Frank-García A, Valdivieso F, Berciano J, et al. (2011). Genetic variation in the tau protein phosphatase-2A pathway is not associated with Alzheimer’s disease risk. BMC Res. Notes 4, 327.
    1. Vilkeviciute A, Kriauciuniene L, Chaleckis R, Deltuva VP, and Liutkeviciene R (2019). COL10A1 (rs1064583) gene variants with predisposition to age-related macular degeneration. Dis. Markers 2019, 5631083.
    1. Wasik U, Schneider G, Mietelska-Porowska A, Mazurkiewicz M, Fabczak H, Weis S, Zabke C, Harrington CR, Filipek A, and Niewiadomska G (2013). Calcyclin binding protein and Siah-1 interacting protein in Alzheimer’s disease pathology: neuronal localization and possible function. Neurobiol. Aging 34, 1380–1388.
    1. Wei M, Brandhorst S, Shelehchi M, Mirzaei H, Cheng CW, Budniak J, Groshen S, Mack WJ, Guen E, Di Biase S, et al. (2017). Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease. Sci. Transl. Med 9, eaai8700.
    1. Wu F, Han B, Wu S, Yang L, Leng S, Li M, Liao J, Wang G, Ye Q, Yuan Z, et al. (2019a). Circular RNA TLK1 aggravates neuronal injury and neurological deficits after ischemic stroke via miR-335-3p/TIPARP. J. Neurosci 39, 7369–7393.
    1. Wu M, Fang K, Wang W, Lin W, Guo L, and Wang J (2019b). Identification of key genes and pathways for alzheimer’s disease via combined analysis of genome-wide expression profiling in the hippocampus. Biophys. Rep 5, 98–109.
    1. Xie Z, Wei M, Morgan TE, Fabrizio P, Han D, Finch CE, and Longo VD (2002). Peroxynitrite mediates neurotoxicity of amyloid beta-peptide1-42- and lipopolysaccharide-activated microglia. J. Neurosci 22, 3484–3492.
    1. Youmans KL, Leung S, Zhang J, Maus E, Baysac K, Bu G, Vassar R, Yu C, and LaDu MJ (2011). Amyloid-β42 alters apolipoprotein E solubility in brains of mice with five familial AD mutations. J. Neurosci. Methods 196, 51–59.
    1. Youmans KL, Tai LM, Nwabuisi-Heath E, Jungbauer L, Kanekiyo T, Gan M, Kim J, Eimer WA, Estus S, Rebeck G, et al. (2012). APOE4-specific changes in Aß accumulation in a new transgenic mouse model of alzheimer disease. J. Biol. Chem 287, 41774–41786.
    1. Young SN (1996). Behavioral effects of dietary neurotransmitter precursors: basic and clinical aspects. Neurosci. Biobehav. Rev 20, 313–323.
    1. Yuan T, Yan F, Ying M, Cao J, He Q, Zhu H, and Yang B (2018). Inhibition of ubiquitin-specific proteases as a novel anticancer therapeutic strategy. Front. Pharmacol 9, 1080.
    1. Zabel M, Schrag M, Crofton A, Tung S, Beaufond P, Van Ornam J, Di-Ninni A, Vinters HV, Coppola G, and Kirsch WM (2013). A shift in microglial β-amyloid binding in Alzheimer’s disease is associated with cerebral amyloid angiopathy. Brain Pathol 23, 390–401.
    1. Zare-shahabadi A, Masliah E, Johnson GVW, and Rezaei N (2015). Autophagy in Alzheimer’s disease. Rev. Neurosci 26, 385–395.
    1. Zhang S, Wang XJ, Tian LP, Pan J, Lu GQ, Zhang YJ, Ding JQ, and Chen SD (2011). CD200-CD200R dysfunction exacerbates microglial activation and dopaminergic neurodegeneration in a rat model of Parkinson’s disease. J. Neuroinflammation 8, 154.
    1. Zhao L, Li Z, Vong JSL, Chen X, Lai HM, Yan LYC, Huang J, Sy SKH, Tian X, Huang Y, et al. (2020). Pharmacologically reversible zonation-dependent endothelial cell transcriptomic changes with neurodegenerative disease associations in the aged brain. Nat. Commun 11, 4413.
    1. Zhu H, Guo Q, and Mattson MP (1999). Dietary restriction protects hippocampal neurons against the death-promoting action of a presenilin-1 mutation. Brain Res. 842, 224–229.

Source: PubMed

3
Subscribe