Page 51 - Read Online
P. 51

Page 346        vonderEmbse et al. Neuroimmunol Neuroinflammation 2020;7:345-59 I  http://dx.doi.org/10.20517/2347-8659.2019.29

               Conclusion: This study demonstrates the exacerbating effects and early manifestation of GxE interactions in
               this model. Furthermore, these findings underscore a period of female-specific vulnerability to epigenetic
               maladaptation during postnatal development, with implications on the faulty later-life adaptability of neuroimmune
               signaling. Further investigation is warranted to evaluate the persistence and relative contribution of these early
               influences on the etiopathology of Alzheimer’s disease.

               Keywords: MicroRNA, microglia, Alzheimer’s disease, developmental origins of adult disease




               INTRODUCTION
               Numerous studies examining the developmental origins of adult disease (DOAD) hypothesis have
               shown that environmental perturbations to neuroimmune development may contribute to later-life
                                                               [1-3]
               neurodegenerative diseases, like Alzheimer’s disease (AD) . The consequence of gene x environment (GxE)
               interactions is much more profound during development due to critical windows in which phenotypic
               plasticity is instructed by, and extremely sensitive to, exogenous signaling. In our previous studies using a
               DOAD model for AD, we reported that the interaction of GxE exacerbated AD susceptibility later in life in
                                                                             [4,5]
               a transgenic AD mouse model postnatally exposed to lead acetate (Pb) . This GxE-related vulnerability
               was concurrent with dysfunctional microglial phenotypes and synaptic defects indicative of atypical
               microglia-neuron interactions, and significantly biased towards females. In agreement with the literature,
               microglia in aging brains can become senescent, dystrophic, and mount ineffective or inappropriate
                                          [6-8]
               responses to neuronal signaling . Importantly, the spatially- and temporally-synchronized development
               of microglia and neurons during the perinatal period fine-tunes this crosstalk necessary for homeostatic
                                 [9]
               signaling in the adult . Thus, imbalances in neuroimmune signaling and function may be developmentally
               promoted.

               Healthy homeostatic interactions between microglia and neurons in adulthood depend on the tightly-
                                                                                                 [9]
               controlled developmental programming of microglia in response to cues from neurons , clearly
               demonstrated in studies of early immune insults resulting in cognitive decline, impaired memory, and
               even neurodegeneration in adulthood [2,10-13] . Mutations to the transmembrane adaptor protein DAP12 or its
                                                                                                       [14]
               ligand TREM2 are associated with a form of presenile dementia called Nasu-Hakola disease in humans ,
               and patients with heterozygous variants of TREM2 have a significantly higher susceptibility to AD [15-17] .
               Furthermore, the typical expression of DAP12 is limited to perinatal microglia and is critical for healthy
               developmental synaptic pruning by microglia [18,19] . DAP12-deficient rodent models exhibit persistent
               synaptic defects to glutamatergic synapses in adulthood [20,21] . Therefore, DAP12 may highlight a particular
               junction of the neuroimmune interface during development by which microglia and neurons establish and
               promote healthy synapse dynamics throughout life, with implications for the promotion of early AD-like
               phenotypes in its absence. However, little is known about the molecular mechanisms through which early-
               life toxicant exposure alters this junction to promote atypical signaling between neurons and microglia that
               then persist beyond development.


               As environmentally sensitive genomic regulators, microRNAs represent a form of long-term epigenetic
               regulation relevant to GxE studies to fine-tune cellular phenotypes. Nearly 60% of all protein-encoding
               genes are thought to be regulated by miRNAs , and, with half-lives nearly 10x longer than mRNA ,
                                                        [22]
                                                                                                       [23]
               numerous studies have already begun exploring the potential of these molecules to act as biomarkers of
               disease . In the healthy adult brain, transient changes to miRNAs are often the result of the immediate
                     [24]
               microenvironment, promoting functional changes to parenchymal targets as a cellular adaption to
               exogenous influences - temporarily shifting the requirements for homeostasis. However, altered miRNAs
               during critical windows of neuroimmune development have direct consequences on the promotion
   46   47   48   49   50   51   52   53   54   55   56