Skip to main page content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2021 Sep;26(9):4687-4701.
doi: 10.1038/s41380-020-0824-3. Epub 2020 Jul 6.

MicroRNA-195 rescues ApoE4-induced cognitive deficits and lysosomal defects in Alzheimer's disease pathogenesis

Affiliations
Free PMC article

MicroRNA-195 rescues ApoE4-induced cognitive deficits and lysosomal defects in Alzheimer's disease pathogenesis

Jiqing Cao et al. Mol Psychiatry. 2021 Sep.
Free PMC article

Abstract

Our recent findings link the apolipoprotein E4 (ApoE4)-specific changes in brain phosphoinositol biphosphate (PIP2) homeostasis to the susceptibility of developing Alzheimer's Disease (AD). In the present study, we have identified miR-195 as a top micro-RNA candidate involved in the ApoE/PIP2 pathway using miRNA profiles in human ROSMAP datasets and mouse microarray studies. Further validation studies have demonstrated that levels of miR-195 are significantly lower in human brain tissue of ApoE4+/- patients with clinical diagnosis of mild cognitive impairment (MCI) or early AD when compared to ApoE4-/- subjects. In addition, brain miR-195 levels are reduced along with disease progression from normal aging to early AD, and cerebrospinal fluid (CSF) miR-195 levels of MCI subjects are positively correlated with cognitive performances as measured by mini-mental status examination (MMSE) and negatively correlated with CSF tau levels, suggesting the involvement of miR-195 in early development of AD with a potential impact on cognition. Similar differences in miR-195 levels are seen in ApoE4+/+ mouse hippocampal brain tissue and cultured neurons when compared to ApoE3+/+ counterparts. Over-expressing miR-195 reduces expression levels of its top predicted target synaptojanin 1 (synj1), a brain PIP2-degrading enzyme. Furthermore, elevating miR-195 ameliorates cognitive deficits, amyloid plaque burden, and tau hyper-phosphorylation in ApoE4+/+ mice. In addition, elevating miR-195 rescues AD-related lysosomal defects in inducible pluripotent stem cells (iPSCs)-derived brain cells of ApoE4+/+ AD subjects while inhibiting miR-195 exacerbates these phenotypes. Together, our data uncover a novel regulatory mechanism of miR-195 targeted at ApoE4-associated brain PIP2 dyshomeostasis, cognitive deficits, and AD pathology.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1. MiR-195 is identified as a top miRNA candidate involved in APOE-regulated synj1 expression.
a Venn diagram shows miR-195 as the only miRNA in common shared among 4 groups: miRNAs differentially expressed between ApoE4+ and ApoE4 carriers in the human ROSMAP dataset, miRNAs differentially expressed between ApoE4+ and ApoE4 in the mouse miRNA array studies, miRNAs negatively correlated with synj1 mRNA in ROSMAP, and miRNAs predicted to target at synj1 mRNA by multiMiR database. Numbers of miRNAs overlapping among subgroups are indicated (red numbers). b Log Fold of changes (LogFC) and p values of differences in miR-195 levels between ApoE4+ and ApoE4 carriers, between female ApoE4+ and ApoE4 carriers in ROSMAP dataset, as well as differences in miR-195 levels between mouse ApoE4+ and ApoE4 treated neurons. c Analysis of correlation between miR-195 and synj1 mRNA in human subjects of the ROSMAP database.
Fig. 2
Fig. 2. Reduction of brain miR-195 levels in human brain and CSF samples is associated with ApoE4 genotype, disease progression, and cognitive decline.
a Amounts of miR-195 (presented as Log2 fold changes) in human parietal cortex tissue of ApoE4+/− subjects (CDR0.5-1) were lower than those in ApoE4−/− subjects. N = 17–18/group; log2FC fold of changes: ApoE4−/− 0.054 ± 0.113 versus ApoE4+/− −0.570 ± 0.178, **p < 0.01 with independent-samples t-tests. b Pattern of reduction in miR-195 levels (presented as Log2 fold changes) along with AD disease progression from normal aging to MCI and early AD. N = 12–19/group; log2FC: 1.626 ± 0.696 in CDR 0 subjects versus 0.242 ± 0.104 in CDR 0.5 MCI patients; versus −0.663 ± 0.135 in CDR 1 AD subjects; *p < 0.05, ****p < 0.0001 with ANOVA tests. c Positive correlation between CSF miR-195 levels and MMSE scores (r = 0.455, p = 0.029; N = 23). d Negative correlation between CSF miR-195 and pTau levels (r = −0.408, p = 0.04; N = 23).
Fig. 3
Fig. 3. MiR-195 expression is reduced in hippocampal brain tissue and cultured primary neurons of ApoE4 mice; modulating miR-195 levels regulates synaptojanin 1 expression.
a Levels of miR-195 were reduced in 12-month old ApoE4 hippocampal brain tissue (log2FC: −0.283 ± 0.069) when compared to those in ApoE3 mice (log2FC: −0.036 ± 0.034). N = 11–13/group with both males and females; **p = 0.0096 with ANOVA tests. A nominal reduction in miR-195 levels was seen in ApoE−/− brains with no statistical significance (log2FC: −0.125 ± 0.067, p = 0.48). b Levels of miR-195 in ApoE−/− neurons treated with ApoE4-CM were reduced (log2FC = −0.314 ± 0.073,) when compared to levels of those treated with ApoE3-CM (log2FC: 0.184 ± 0.094). N = 5/group; **p = 0.003 with independent-samples t-tests. c Differences in miR-195 expression levels between ApoE3-CM and ApoE4-CM treated neurons were abolished in the presence of RAP. The treatment of RAP in the presence of ApoE3-CM led to a reduction in miR-195 levels (log2FC: −1.648 ± 0.125; p < 0.0001), whereas in ApoE4-CM treated conditions, miR-195 levels were much lower at baseline with a trend of improvement in the presence of RAP treatment (ApoE4 CM + BSA log2FC: −3.193 ± 0.144 versus ApoE4 CM + RAP log2FC: −2.678 ± 0.054; p = 0.052). N = 3/group; ****p < 0.0001 by One-Way ANOVA tests. d Synj1 protein levels were reduced with miR-195 over-expression but not miR-374 over-expression in ApoE−/− hippocampal neurons in the presence of ApoE4-CM. N = 4/group; synj1 levels with miR-195: 62.87 ± 4.48% of controls, **p = 0.001; with miR-374: 102.4 ± 7.77% of controls, p = 0.93.
Fig. 4
Fig. 4. Over-expression of miR-195 rescues cognitive deficits and ameliorates AD-associated pathologies in ApoE4 mouse models.
a Preference index = (time exploring novel object)/(time exploring novel object + time exploring familiar object) and discrimination index = (time exploring novel object- time exploring familiar object)/(time exploring novel object + time exploring familiar object) in 4 groups of mice: ApoE4+/+ scramble injection, ApoE4+/+ miR-195 injection, ApoE3+/+ scramble injection, and ApoE3+/+ miR-195 injection. N = 19–23/group with both males and females; *p < 0.05 with ANOVA tests. b Levels of pTau in KI mouse hippocampus. N = 8/group with both males and females; *p < 0.05; **p < 0.01 with ANOVA tests. c Preference index and discrimination index in 8 groups of mice: ApoE4+/+ FAD male scramble injection, ApoE4+/+ FAD male miR-195 injection, ApoE4+/+ FAD female scramble injection, ApoE4+/+ FAD female miR-195 injection, ApoE3+/+ FAD male scramble injection, ApoE3+/+ FAD male miR-195 injection, ApoE3+/+ FAD female scramble injection, and ApoE3+/+ FAD female miR-195 injection. N = 6–10/group; *p < 0.05 with ANOVA tests. Levels of d pTau and e oligomer Aβ42 in EFAD mouse hippocampus. N = 6/group with both males and females; *p < 0.05; ***p < 0.001; ****p < 0.00001 with ANOVA tests. f Amyloid plaque burden in EFAD mouse hippocampus. A representative image of brain section is shown. Amyloid plaque load is quantified by density measured as area of plaques per mm2 of brain region, as well as total numbers of plaques/μm2 in 4 groups of mice: ApoE4+/+ FAD scramble injection, ApoE4+/+ FAD miR-195 injection, ApoE3+/+ FAD scramble injection, and ApoE3+/+ FAD miR-195 injection. N = 3/group; *p < 0.05; **p < 0.01 with ANOVA tests.
Fig. 5
Fig. 5. Over-expression of miR-195 rescues lysosomal defects in ApoE4 iPSC-derived brain cells.
Representative immunofluorescence co-staining of lysosomes (Lysotracker: red fluorescence), a neuronal marker MAP-2 (green fluorescence) and DAPI (blue fluorescence) of iPSC-derived neuron and astrocyte co-culture of (a) ApoE3+/+ normal aging (NA) and (b) ApoE4+/+ AD subjects with various conditions: scramble control (ctrl), miR-195, and miR-195 inhibitor (miR inh). Alternatively, ApoE3+/+ and ApoE4+/+ N = 3–6/conditions. c Quantification of all lysosomes by size (measured by areas; μm2) of 60–90 neurons (MAP-2+) in each experimental condition, the distribution of lysosome sizes/cell (measured by diameters; 0–10 μm, 10–20 μm, 20–30 μm and >30 μm), as well as the number of lysosomes in each cell (grouped by 1–5, 6–10, 11–15, 16–20, and >20 lysosomes/cell). ****p < 0.00001 with ANOVA tests.

Similar articles

Cited by 9 articles

References

    1. Mayeux R. Epidemiology of neurodegeneration. Annu Rev Neurosci. 2003;26:81–104. - PubMed
    1. Fagan AM, Watson M, Parsadanian M, Bales KR, Paul SM, Holtzman DM. Human and murine ApoE markedly alters A beta metabolism before and after plaque formation in a mouse model of Alzheimer’s disease. Neurobiol Dis. 2002;9:305–18. - PubMed
    1. Jiang Y, Sun XC, Gui L, Tang WY, Zhen LP, Gu YJ, et al. Lack of association between apolipoprotein E promoters in epsilon4 carriers and worsening on computed tomography in early stage of traumatic brain injury. Acta Neurochir Suppl. 2008;105:233–6. - PubMed
    1. Jordan J, Galindo MF, Miller RJ, Reardon CA, Getz GS, LaDu MJ. Isoform-specific effect of apolipoprotein E on cell survival and beta-amyloid-induced toxicity in rat hippocampal pyramidal neuronal cultures. J Neurosci: Off J Soc Neurosci. 1998;18:195–204. - PMC - PubMed
    1. Ma J, Yee A, Brewer HB, Jr, Das S, Potter H. Amyloid-associated proteins alpha 1-antichymotrypsin and apolipoprotein E promote assembly of Alzheimer beta-protein into filaments. Nature. 1994;372:92–4. - PubMed

Publication types