Next Article in Journal
Serum Oxidative Status in People with Obesity: Relation to Tissue Losses, Glucose Levels, and Weight Reduction
Next Article in Special Issue
Betula pendula Leaf Extract Targets the Interplay between Brain Oxidative Stress, Inflammation, and NFkB Pathways in Amyloid Aβ1-42-Treated Rats
Previous Article in Journal
The Coumarin-Derivative Esculetin Protects against Lipotoxicity in Primary Rat Hepatocytes via Attenuating JNK-Mediated Oxidative Stress and Attenuates Free Fatty Acid-Induced Lipid Accumulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Contribution of Hippocampal All-Trans Retinoic Acid (ATRA) Deficiency to Alzheimer’s Disease: A Narrative Overview of ATRA-Dependent Gene Expression in Post-Mortem Hippocampal Tissue

1
Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
2
Department of Pharmacology and Neuroscience and Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
3
Department of Pharmacology and Neuroscience, Garrison Institute on Aging, Center of Excellence for Translational Neuroscience and Therapeutics, and Center of Excellence for Integrated Health, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
*
Author to whom correspondence should be addressed.
Antioxidants 2023, 12(11), 1921; https://doi.org/10.3390/antiox12111921
Submission received: 31 August 2023 / Revised: 4 October 2023 / Accepted: 5 October 2023 / Published: 27 October 2023
(This article belongs to the Special Issue Oxidative Stress and Alzheimer’s Disease)

Abstract

:
There is accumulating evidence that vitamin A (VA) deficiency contributes to the pathogenesis and progression of Alzheimer’s disease (AD). All-trans retinoic acid (ATRA), a metabolite of VA in the brain, serves distinct roles in the human hippocampus. Agonists of retinoic acid receptors (RAR), including ATRA, promote activation of the non-amyloidogenic pathway by enhancing expression of α-secretases, providing a mechanistic basis for delaying/preventing amyloid beta (Aβ) toxicity. However, whether ATRA is actually deficient in the hippocampi of patients with AD is not clear. Here, using a publicly available human transcriptomic dataset, we evaluated the extent to which ATRA-sensitive genes are dysregulated in hippocampal tissue from post-mortem AD brains, relative to age-matched controls. Consistent with ATRA deficiency, we found significant dysregulation of many ATRA-sensitive genes and significant upregulation of RAR co-repressors, supporting the idea of transcriptional repression of ATRA-mediated signaling. Consistent with oxidative stress and neuroinflammation, Nrf2 and NfkB transcripts were upregulated, respectively. Interestingly, transcriptional targets of Nrf2 were not upregulated, accompanied by upregulation of several histone deacetylases. Overall, our investigation of ATRA-sensitive genes in the human hippocampus bolsters the scientific premise of ATRA depletion in AD and that epigenetic factors should be considered and addressed as part of VA supplementation.

Graphical Abstract

1. Introduction

Approximately 6.7 million Americans are currently living with Alzheimer’s disease (AD) [1]. As the aging US population grows, 13.8 million Americans are projected to have AD by 2060 [1]. Currently, two treatments for AD are approved by the Food and Drug Administration, but there are no cures that are capable of reversing or halting the neurodegenerative progression of AD. Genetic predisposition and age are unmodifiable risk factors for AD. However, several environmental and lifestyle risk factors, such as diet, smoking, and physical activity, are modifiable. Poor diet is a leading cause of death in the US and worldwide [2,3], and is associated with a number of comorbidities (e.g., cardiovascular disease, obesity, diabetes) that increase the risk for AD [4,5,6,7,8]. Observational studies and clinical trials have demonstrated the protective benefits of a healthy diet in relation to AD pathogenesis [9,10]. Moreover, dietary intake of vegetables is associated with high levels of serum antioxidants (AOs) and reduced Aβ accumulation [11].
It is widely accepted that the toxic accumulation of amyloid beta (Aβ) and neurofibrillary tangles leads to progressive cognitive decline, ultimately culminating in AD-induced death [12]. The Mitochondrial Free Radical Theory of Aging proposes that reactive oxygen species (ROS), produced as a product of normal mitochondrial metabolism, cause oxidative damage to proteins, lipids, and nucleic acids which accumulates across the lifespan [13]. Antioxidants (AOs), both diet-derived, and Nrf2-mediated endogenous AOs, help prevent ROS-induced damage. The condition in which cellular AO capacity is insufficient to neutralize ROS is termed oxidative stress (OS). Therefore, the preconditions necessary for ROS-induced oxidative damage require not only ROS generation, but also the depletion of dietary and/or endogenous AO defenses. AO replenishment is an intuitive therapeutic strategy to attenuate, delay, or altogether prevent age-related cognitive decline (ARCD) and AD. Paradoxically, the use of generic ROS scavengers, such as vitamin E, as AO monotherapies have failed in clinical trials [14,15,16]. These failures have revealed that faulty assumptions and major knowledge gaps exist in our models of the biological etiology and mechanisms of AD.
We hypothesize that the progression of AD may involve deficiencies in specific AOs that have roles beyond that of generic ROS scavengers. In this study, we highlight the multifaceted roles of all-trans retinoic acid (ATRA) with particular emphasis on the growing evidence of ATRA deficiency in AD. Moreover, we present transcriptomic evidence of ATRA deficiency in human AD reflecting its in vivo roles as both a transcription factor and an AO.

2. Results and Discussion

2.1. ATRA Transport and Metabolism

Under healthy physiological conditions, ATRA sufficiency promotes health through the scavenging of excess ROS and by inducing the expression of genes critical for synaptic function and neuroprotection (Figure 1A). Retinol (a form of VA) is first transported from liver stores to the hippocampus through the bloodstream, bound to a carrier complex that consists of retinol binding protein 4 (RBP4) [17] and transthyretin (TTR) [18]. Retinol crosses the blood brain barrier, facilitated via RBP4 binding to its receptor, signaling receptor and transporter of retinol STRA6 (STRA6) [19]. Retinol also enters neurons via STRA6, which catalytically dissociates retinol from RBP4 and transports it across the plasma membrane [19]. Once within the cytoplasm, retinol binds to cellular retinol binding protein 1 (RBP1) and is converted into retinal by one of several retinol dehydrogenase enzymes (RDH). RBP1-bound retinal is then converted into ATRA by a retinaldehyde dehydrogenase enzyme (ALDH1a1/2/3) and is transferred to several binding proteins: cellular retinoic acid binding protein 1/2 (CRABP1/2) or fatty acid binding protein 5 (FABP5). As shown in cell lines, the binding of ATRA to either CRABP1/2 or FABP5 is a defining step because it determines nuclear receptor localization and, subsequently, which genes are transcriptionally activated [20,21,22].

2.2. Cellular Binding Proteins Traffic ATRA to Distinct Nuclear Receptors, Collectively Promoting Activation of the Nonamyloidogenic Pathway

When bound to CRABP2, ATRA is transported into the nucleus and transferred to retinoic acid receptor (RARs), as has been shown in astrocytes [23,24]. ATRA-RARα becomes heterodimerized with the retinoid X receptor (RXR). The ATRA-RARα/RXR complex enables activation of transcription, as RXR is a non-permissive partner [25]. However, RXR can also bind many co-agonists, including docosahexaenoic acid (DHA), 9-cis-RA, and others [26,27,28]. ATRA binding to RARα/RXR results in the dissociation of bound co-repressors and recruits transcriptional activators and histone acetyltransferases (HATs). This heterodimer binds to promoter retinoic acid response elements (RAREs) to increase the transcription of ATRA-responsive genes, including the α-secretase ADAM10 [29] which mediates processing of amyloid precursor protein (APP) into non-amyloidogenic fragments, as well as additional genes involved in synaptic plasticity that are neuroprotective against AD pathogenesis. Unlike the toxic amyloid-β peptides that result from β-secretase (BACE1) cleavage, these cleavage products do not aggregate to form plaques or catalyze the aberrant formation of ROS (Figure 1A).
When bound to FABP5, ATRA is transported into the nucleus and transferred to peroxisome proliferator-activated receptor β/δ (PPARβ/δ), which also dimerizes with RXRs [30]. The PPARβ/δ-RXR heterodimer then binds to peroxisome proliferator response elements (PPREs) to express genes important for neuroprotection [31,32,33,34] (Figure 1A). ATRA binding to PPARβ/δ-RXR induces transcription of genes that function to minimize OS, neuroinflammation, and neurotoxicity, and to decrease Aβ and hyperphosphorylated tau (p-tau) levels [30,35,36,37,38]. PPARβ/δ knockout mice exhibit increased expression of β-secretase (BACE1), receptor for advanced glycation endproducts (RAGE), proinflammatory and proapoptotic mediators, and tau hyperphosphorylation [30,35], implicating PPARβ/δ receptors in AD pathology. Mice treated with GW0742, a selective PPARβ/δ agonist, showed a reduction in parenchymal Aβ plaque deposition, diminished neuroinflammatory and apoptotic states, and decreased activation of microglia [39,40,41]. Similarly, T3D-959, a synthetic agonist of both PPARβ/δ and PPARγ, with 15-fold higher selectivity for PPARβ/δ, has shown promise in clinical trials [42,43,44]. Other PPARs outside of PPARβ/δ, such as PPARα and PPARγ, have also been linked to neuroprotection against AD [45,46].
Similarly to the ATRA-mediated activation of RARα, the activation of PPARα promotes the non-amyloidogenic processing of APP by inducing the transcription of the α-secretase ADAM10 [47,48,49]. Activation of PPARγ results in decreased levels of neuroinflammation, OS, and Aβ; however thiazolidinedione drugs which are agonists for PPARγ have not shown promise in clinical trials due to poor BBB penetration [50]. Interestingly, both PPARα and PPARβ/δ are downregulated in AD brains, whereas PPARγ is upregulated [51]. Although PPARα and PPARγ are not directly activated by ATRA, their dimer partner RXR is activated by 9-cis-retinoic acid [52,53], an isomerization product of ATRA, suggesting a mechanism by which ATRA and/or retinol levels may affect these pathways.
Transcription of RARα- and PPARβ/δ-sensitive genes is dependent on the levels of CRABP2 and FABP5, respectively [21,22]. Increased expression of CRABP2 yields higher transcription of RARα-regulated genes, whereas increased expression of FABP5 yields increased expression of PPARβ/δ-regulated genes [21,54]. ATRA has a similar binding affinity for CRABP2 and FABP5, though the agonism depends on competing fatty acids [20]. Therefore, many factors depend on how the two pathways are tuned to be differentially responsive to different ATRA concentrations, suggesting that the key regulatory activities of each pathway are relevant under distinct physiological states [31]. Additionally, PPARβ/δ polymorphisms in patients do not seem to be significant risk factors for AD, arguing against a central role of ATRA-PPARβ/δ in brain neuroprotection [55,56]. In summary, ATRA-responsive RARα and PPARβ/δ pathways appear to converge on mechanisms which reduce OS and Aβ accumulation, which should, in principle, delay AD onset and progression. If ATRA concentrations remain too low for significant binding interactions to occur, then PPARβ/δ polymorphisms would not be expected to influence AD progression. However, PPARβ/δ agonists may still be able to provide neuroprotective benefits in patients treated with PPARβ/δ agonists or retinol supplementation.

2.3. The Role of ATRA in AD Animal Models

Our current understanding of the role ATRA in the hippocampus of rodent AD models come from six lines of converging evidence. First, VA plays an important role in hippocampal-dependent learning. VA deficiency in the brain is associated with impairment in hippocampal-dependent learning [57,58,59,60]. In contrast, ATRA sufficiency stimulates neurogenesis, hippocampal dendritic growth, and hippocampal learning and memory operations [57,61,62]. Second, ATRA deficiency increases Aβ levels in the normal rat brain by decreasing expression of the α-secretase enzyme ADAM metallopeptidase domain 10 (ADAM10), disrupting the balance between non-amyloidogenic and amyloidogenic APP processing pathways [63,64]. Moreover, direct activation of RARs upregulates ADAM10 and promotes activation of the non-amyloidogenic pathway, reducing Aβ generation and deposition [65,66,67]. Third, ATRA or retinol supplementation can restore hippocampal ATRA levels, preserve learning capability, rescue hippocampal-dependent learning impairment, and reduce amyloid load in AD mouse models [57,68,69]. Fourth, even when liver stores of VA are sufficient, hippocampal ATRA levels are dramatically reduced with hepatic inflammation and/or age, suggesting liver-brain dysregulation in the hepatic release, vascular transport, or hippocampal processing of VA [57,59,70]. Fifth, chronic neuroinflammation in microglia upregulates the expression of CYP26, an enzyme involved in ATRA degradation, implying that chronic neuroinflammation, as observed in AD, is associated with reduced hippocampal ATRA levels [71]. Treatment with ATRA or a RARα agonist exerts potent anti-inflammatory effects [68,71,72,73]. Finally, ATRA plays a non-genomic role in homeostatic plasticity by balancing excitatory and inhibitory synaptic strength, mediated by postsynaptic calcium-dependent regulation of ATRA synthesis [74,75,76,77]. In contrast, RARα conditional knockout mice, which are deficient in ATRA signaling in the hippocampal CA1 region, display runaway Hebbian synaptic plasticity [78].

2.4. The Role of ATRA in Human AD

There is growing evidence for a role of ATRA in the human hippocampus with regards to cognitive function [79]. First, there is a negative correlation between retinol/carotenoid intake or serum levels and AD-related cognitive decline [11,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94]. Second, there is a negative correlation between serum retinol/carotenoid levels and biomarkers of OS [95,96]. On the other hand, a positive correlation has been shown between serum carotenoids levels and telomere length in leukocytes, which is considered a biomarker of heathy aging [84,97]. Third, genes involved in retinol metabolism and function have been linked to late-onset AD [98,99,100]. Moreover, loss of function mutations in the α-secretase ADAM10 result in seizures, further strengthening the association between AD and epilepsy [101,102]. Consistent with this association, ADAM10 overexpression suppresses seizures and inflammation in epilepsy animal models [103]. Fourth, VA/ATRA dysregulation is associated with environmentally induced comorbidities that are now recognized as leading risk factors for AD, including cardiovascular disease, obesity, hepatosteatosis, and diabetes [104,105]. Lastly, a randomized control trial demonstrated that treatment with the RAR agonist isotretinoin increased activation of the nonamyloidogenic pathway, as indicated by increased cerebrospinal fluid levels of APPsα, a neuroprotective cleavage product of APP [106].

2.5. A Critical Knowledge Gap: ATRA-Sensitive Gene Expression in Postmortem Human AD Hippocampus

Despite the considerable amount of correlative evidence for ATRA deficiency in AD patients and accompanying hippocampal-related learning deficits, there is currently no direct evidence that ATRA is deficient in the hippocampus of AD patients. We surmised that hippocampal ATRA deficiency could be inferred through the dysregulation of ATRA-sensitive genes, essentially making use of ATRA-sensitive genes as endogenous ATRA sensors (Figure 2). Therefore, using a previously published RNAseq dataset from the hippocampi of postmortem AD and control brains [107], we performed a secondary analysis of publicly available hippocampal transcriptomic data that was generated from 20 AD cases and 10 controls, originally acquired from the Netherlands Brain Bank by van Rooij and colleagues [107]. This data set contains a heterogeneous population of AD hippocampal tissue samples that included both sexes and multiple APOE genotypes [107]. We examined genes known to possess RAREs and/or known to be transcriptionally controlled by ATRA (the VA transcriptome) [108,109,110,111], which included 297 genes that were shown to possess RARβ binding sites [112]. In addition, we also consulted the web-accessible transcription factor databases TF2DNA [113] and Enrichr [114,115,116] for ATRA-sensitive genes and ATRA-related transcription factors.
Collectively, this study provides new evidence that numerous ATRA-sensitive genes are differentially expressed (DE) in AD compared to control hippocampus. We discovered that RAR co-repressors were upregulated, suggesting that, in addition to ATRA deficiency, downstream ATRA-responsive transcription is actively blocked. We also found indirect evidence for ATRA depletion via Nrf1 and Nrf2, master OS-responsive transcription factors. Nrf1 and Nrf2 were upregulated in the hippocampus of human AD brains, consistent with ATRA depletion and its activity as an antioxidant. Interestingly, we discovered that transcriptional targets of Nrf2 were downregulated, providing evidence that even though Nrf2 was upregulated in AD hippocampus, this transcription factor was unable to successfully induce the expression of AO defenses. Finally, we found that several histone deacetylases (HDACs) were upregulated. This finding, along with the finding that Nrf2-responsive genes were downregulated despite Nrf2 upregulation, suggests that epigenetic silencing may interfere with the capability of transcription factors such as RAR and Nrf2 to transactivate their target genes, resulting in the depletion of both exogenous and endogenous antioxidants and collectively contributing to oxidative stress and neurodegeneration. In this review, we discuss some of the transcriptomic findings supporting the dysregulation of retinoid signaling in the hippocampus of AD patients.

2.6. Genes Mediating Retinol Transport, ATRA Synthesis, and ATRA Metabolism Are Dysregulated in Hippocampal Tissue from Post-Mortem AD Brains

We first examined the expression of genes involved in retinoid transport and metabolism, as well as retinoid responsive transcription factors (Table 1). Retinol binding protein 4 (RBP4) and transthyretin (TTR) are retinol carriers produced by the liver (Figure 1). Interestingly, RBP4 and TTR transcripts were detected in postmortem hippocampal tissue. Moreover, RBP4 was significantly downregulated in human AD. If hippocampal RBP4 transcript is reflective of the level of RBP4 protein in circulation, the dysregulation of hippocampal RBP4 transcript may signify changes in liver RBP4 expression, suggesting impaired retinol transport along the liver-brain axis. Alternatively, brain and liver RBP4 transcript expression may be completely independent of each other; future research is needed to determine relationships in RBP4 expression between liver and brain. RBP1 (frequently designated CRBP1), the intracellular retinol carrier that accepts retinol from the STRA6 transporter [20], was also found to be transcriptionally downregulated in the hippocampus of AD brains (Table 1). Both RBP1 and RBP4 are known to be transcriptionally regulated by ATRA [108,110,111,117,118]. Retinol dehydrogenase 12 (RDH12) is one of the major cytoplasmic enzymes used in the conversion of retinol to retinal and was significantly downregulated. ADLH1A1, ADLH1A2, and ADLH1A3 are involved in the cytoplasmic conversion of retinal to ATRA. ALDH1A3 was significantly downregulated, suggesting reduced production of ATRA. CRABP2 transfers ATRA to RARα, whereas FABP5 transfers ATRA to PPARβ/δ [31]. CRABP2 and FABP5 transcripts were both detected in the hippocampus but not significantly dysregulated.
CYP26A1 and CYP26B1 transcripts encode ATRA-metabolizing enzymes. CYP26A1 is known to contain canonical DR5 RARE motifs within its promoter region [111,119], enabling feedback control of ATRA levels. CYP26A1 and CYP26B1 cause hydroxylation of ATRA, which promotes the clearance of ATRA used for intracellular and paracrine homeostatic balancing, as well as the formation of other bioactive retinoids that bind to RAR isoforms with lower but variable affinities: 4-OH-RA, 18-OH-RA, 16-OH-RA, and 4-oxo-RA [120,121]. CYP26A1 and CYP26B1 transcripts were significantly downregulated in AD hippocampus. One simple interpretation of the downregulation of these CYP26 enzymes is that it is likely reflective of ATRA deficiency, as both are induced by ATRA through feedback regulation to protect neurons against excessive accumulation of ATRA [120,122].
Expression levels of several RARs (RARA, RARB) were not significantly changed in the AD condition. However, the interpretation is complicated by the fact that both RARA and RARB have distinct isoforms with different transcription start sites, some of which are directly regulated by RAREs, while some are regulated through indirect means. For example, the promoter sequence regulating the transcripts encoding the RARα1 isoform lacks a RARE, while the promoter regulating expression of the RARα2 isoform does contain a RARE [123,124]. Unfortunately, RNA-sequencing results do not discriminate between these two transcripts. RARG was upregulated. However, RARG is unconventional in that the RARE in the promoter is flanked by SP1 transcription factor binding sites [125]. SP1 has been shown to be upregulated in AD [126], which is also the case here (Table 1). Overall, despite these caveats, we interpret these observations to be generally indicative of ATRA deficiency.

2.7. Transcriptional Upregulation of RAR-Related Co-repressor Genes in the Post-mortem Hippocampus in AD

In the absence of ATRA, RAREs are normally repressed by a number of RAR co-repressors, thereby interfering with the transcription of ATRA-sensitive genes [127]. We detected the RAR-related co-repressors NCOR1, NCOR2, ZBTB16, TNIP1, RIF1, and LCOR in postmortem AD hippocampal tissue. Importantly, most of these co-repressors [127,128] were significantly and uniformly upregulated (Table 2), consistent with impaired transcription of RAR-dependent downstream signaling. Nuclear Receptor Corepressor 1 (NCOR1) is involved in ATRA-independent transcriptional repression [129], which complexes with histone deacetylases (HDACs) to promote condensation of chromatin and epigenetic silencing. Similarly, promyelocytic leukemia zinc finger protein (PLZF, currently known as ZBTB16) complexes with RARα, resulting in transcriptional repression likely by preventing RXR-RARα heterodimerization [130,131]. However, ZBTB16 also represses the transcriptional activity of other Class II (RXR heterodimer) nuclear receptors, so it is not restricted to RXR-RARα [131]. Interestingly, TNIP1 is also a co-repressor of RARα and PPARs. However, it atypically represses these nuclear receptors when they are bound to ATRA [128,132]. This observation suggests that TNIP1 upregulation may interfere with ATRA-sensitive transcription of RAR genes [128,132]. RIF1 has also been shown to have RAR co-repressor activity, but it is not clear to what extent it has co-repressor activity at other nuclear receptors [133]. We detected a modest upregulation of RIF1, though the significance did not meet the false positive rate threshold. Finally, LCOR is a co-repressor of RARs and other types of nuclear receptors [134], which was significantly upregulated. Generally, the upregulation of RAR-related co-repressors is consistent with transcriptional block of RAR signaling in human AD, corroborating the transcriptional downregulation of ATRA-sensitive genes, collectively converging on a strong scientific premise for ATRA deficiency.

2.8. Increased Expression of OS and Neuroinflammatory Genes Is Consistent with ATRA Depletion

Depletion of antioxidants in AD are likely to shift redox balance toward elevated OS [135,136,137]. In addition to being a hormone-like nuclear receptor [138,139], ATRA, as well as Pro-VA carotenoids and retinol, is thought to possess intrinsic antioxidant/ROS scavenging properties [140,141,142]. Therefore, we hypothesize that enhancement of OS biomarkers, as well as compensatory increases in endogenous AO defenses, should accompany the downregulation of ATRA-sensitive genes. We found significantly upregulated transcription of the major OS-responsive transcription factors Nrf1 (NFE2L1) and Nrf2 (NFE2L2). Nrf1/2 bind to antioxidant response elements (AREs) to activate enzymes and molecules involved in endogenous AO defenses [141]. To be consistent with Nrf2-mediated gene expression, these findings should be accompanied by downstream increases in the expression of Nrf1/2 target genes. Interestingly, we found a significant downregulation in transcripts of Nrf2-regulated endogenous AO defense genes (Table 3), including NADPH Quinone oxidoreductase enzyme 1 and 2 (NQO1 and NQO2), glutathione s-transferase alpha 4 (GSTA4), glutathione s-transferase mu 4 (GSTM4), prostaglandin reductase 1 (PTGR1), heme oxygenase 2 (HMOX2), superoxide dismutase type 1 (SOD1), and glutaredoxin (GLRX). These results are consistent with the hypothesis that aberrant AD-related epigenetic silencing may limit the ability of transcription factors such as Nrf2 or RAR/RXR to initiate and/or maintain transcription. This downregulation of Nrf2-mediated endogenous AO defenses was not generally uniform, with catalase (CAT) transcription being modestly upregulated. This observation may indicate that epigenetic silencing in AD is not a uniform process, or that CAT upregulation could have been driven by other transcription factors which act at the promoter of the CAT gene [143]. This latter scenario seems unlikely—it is difficult to envision how silenced promoters would maintain selective responsiveness to only certain transcription factors.
Together with Nrf1 and Nrf2, the family of homodimeric and heterodimeric transcription factors known as Nuclear Factor Kappa B (NF-κB) regulates and coordinates cellular responses to OS and neuroinflammation. We found that the NF-κB family genes NFKB1 (also called p105) and NFKB2 (also called p52) were upregulated in the hippocampus of AD patients (Table 3), indicative of neuroinflammation and OS seen in the AD condition [144,145]. Proinflammatory cytokines such as Tumor Necrosis Factor-Alpha (TNF, commonly called TNF-α) and Interleukin 1 Beta, (IL1B, commonly called IL-1β), present at elevated levels in the blood of the AD patients, are activators of NF-κB [146]. RELA (also known as p65, also a transcription factor of the NF-κB family), transcriptionally represses ARE-sensitive gene expression by competition with Nrf2 for binding of the CBP/p300 co-activator and by recruiting HDAC3 to AREs [147]. This prevents the production of endogenous antioxidants, further activating NF-κB in a potentially pathological positive-feedback cycle [147]. Furthermore, NF-κB can translocate Keap1 into the nucleus to facilitate Nrf2 degradation and can even regulate Nrf2 expression by binding to its promoter region [148]. On the other hand, Nrf2 regulates NF-κB activity by reducing levels of ROS and inhibiting RAC1-mediated activation of NF-κB [148]. Moreover, studies have shown that ATRA counteracts neuroinflammation by reducing pro-inflammatory cytokines and modulating NF-κB activation, suggesting that upregulation of NF-κB transcription factors, and possibly neuroinflammation in general, are promoted by ATRA deficiency [149,150,151,152,153,154,155,156,157].

2.9. Crosstalk between RAR and Nrf2 Signaling

As part of homeostatic balance in the healthy brain, redox balance should be achieved by a combination of exogenous and endogenous antioxidants. Therefore, Nrf2- and RAR signaling pathways should have mechanisms for cross-sensing their activation states. Nrf2 signaling and AO defenses are sensitive to oxidative stress, which we argue may be accompanied by depleted ATRA availability. However, sufficient bidirectional control mechanisms should be in place such that sufficient ATRA bioavailability suppresses Nrf2 signaling in the nucleus by trapping Nrf2 in the cytoplasm through Keap1-Nrf2 interactions, facilitating Nrf2 degradation (Figure 3A). Indeed, there is also evidence that Nrf2 can also be directly sequestered by ATRA-liganded RARα by binding to the Neh7 domain on Nrf2, thereby reducing transcription of Nrf2-mediated AO defenses [158,159]. Similar to RARα agonists, RXRα agonists can repress ARE-dependent gene expression [160]. Moreover, the minimization of ROS accumulation, contributed by ATRA sufficiency, facilitates Keap1-Nrf2 association, which inactivates downstream actions of Nrf2 through subsequent ubiquitination and proteasomal degradation [161]. Interestingly, RXRα has also been shown to sequester Nrf2 without needing to be liganded to ATRA or heterodimerized to RARα [162]. Additionally, it has been shown that in acute myeloid leukemia (AML) cells, ATRA suppress Nrf2 translocation into the nucleus by an unspecified mechanism, further repressing Nrf2-mediated effects [163]. Moreover, ATRA may even directly prevent Nrf2 translocation into the nucleus [163,164], which indicates a complex relationship between exogenous AO sources (i.e., ATRA) and endogenous AOs in maintaining redox balance (Figure 3A). However, further study is required to confirm that this response occurs with neuronal Nrf2. Together, these findings suggest that RAR- and Nrf2-mediated signaling are in homeostatic equilibrium, in which redox balance is in part determined by ATRA availability and cellular expression of Nrf2-mediated endogenous AO defense mechanisms. With ATRA sufficiency, multiple mechanisms exclude Nrf2 from entering the nucleus through direct binding of liganded RARα as well as through Nrf2-Keap1 interactions, ultimately resulting in repressed expression of Nrf2-mediated endogenous AOs. This is not to suggest a complete deactivation of Nrf2-induced expression, rather a coordinated attenuation dependent on hippocampal ATRA concentration which modulates cellular antioxidant mechanisms to counteract ROS accumulation.
By contrast, ATRA deficiency or RARα antagonism promotes Nrf2 binding to AREs [159]. Moreover, the resulting increase in OS mediated by ATRA depletion leads to the dissociation of Nrf2 from Keap1 (Figure 3B). This mechanism occurs via oxidation of the thiol groups (-SH) of cysteine residues on Keap1 by ROS, facilitating the release and translocation of the transcription factor Nrf2 from the cytosol to the nucleus [165]. Nrf2 subsequently dimerizes with small Maf (sMaf) and binds to AREs, inducing the upregulation of endogenous AO pathways that counteract ROS accumulation and limit further oxidative damage to cellular machinery [166,167,168]. Given this intimate association between RARα and Nrf2 signaling, the dysregulation of ATRA-sensitive genes combined with the engagement of Nrf2-sensitive endogenous AO defenses corroborates the idea of ATRA deficiency. In some circumstances, the mechanism of ATRA-induced downregulation of Nrf2 signaling could exacerbate oxidative damage. In the context of leukemia therapy, dismantling AO defenses through ATRA-induced downregulation of Nrf2 signaling can be exploited to help kill cancer cells [163].
We propose that the bioavailability of ATRA and endogenous AOs work in a coordinated fashion to maintain a homeostatic equilibrium of redox balance in the hippocampus. However, in aging and AD, this interplay between ATRA- and Nrf2-mediated regulation may be complicated by epigenetic changes that include changes in histone acetylation/deacetylation balance and RARE/PPARE/ARE related transcription, rendering RAREs, PPAREs, and AREs progressively inaccessible to the RAR-, PPAR- and/or Nrf2-mediated transcription needed to finely tune redox balance within memory circuits (Figure 4A). The eventual homeostatic collapse of this redox balance may hasten AD onset and progression (Figure 4B).

2.10. Dysregulation of Mitochondria-Related Genes Is Consistent with ATRA Depletion, Increased Oxidative Stress, and Mitochondrial Aβ Accumulation

A number of genes involved in mitochondrial (mt) function (TOMM20, TOMM40, OPA1, DNM1L) were significantly downregulated in AD in the human hippocampus (Table 3), suggestive of mt dysfunction induced by OS. Translocase of outer mitochondrial membrane 20 (TOMM20) and 40 (TOMM40) exist at the outer mitochondrial membrane and function to import mitochondrial preproteins synthesized within the cytosol [169,170,171]. Both TOMM20 and TOMM70 have previously been shown to be decreased in the AD hippocampus, indicative of mitochondrial respiration dysfunction [172,173]. The TOMM import machinery is also used in the translocation and accumulation of Aβ into the mitochondrial cristae, which is associated with synaptic deficits [174,175,176]. Therefore, the downregulation of the TOMM proteins may be attributed to active protective mechanisms in addition to transcriptional inactivity due to the absence of agonist. Optic atrophy 1 (OPA1) exists in the inner mitochondrial membrane (IMM) and normally functions in the fusion of the IMM [177]. OPA1 also plays a role in combating ROS accumulation and is similarly decreased in hippocampal AD brains [178,179].
Finally, dynamin-1-like protein (DNM1L; DRP1) is a GTPase that promotes mitochondrial fission, distribution of mitochondria throughout the neuron, neuronal differentiation, synapse formation, and dendrite formation [180,181,182]. ATRA has recently been shown to upregulate DNM1L protein expression [183]. Interestingly, DNM1L mRNA downregulation in the AD hippocampus is correlated with upregulation of retinoic acid receptor-related orphan receptor alpha (RORA) mRNA [184]. In accordance with this study, we observed that DNM1L mRNA was significantly downregulated and RORA mRNA upregulated in the human AD hippocampus (Table 3). The underlying molecular mechanisms are unclear because unlike RORβ, RORα is not activated by ATRA [185]. Furthermore, multiple studies allude to increased expression of DNM1L as a causative factor in AD pathogenesis due to increased mitochondrial fragmentation; beneficial effects of DNM1L inhibition have been observed in AD models [186,187,188,189]. Other studies have proposed that Aβ and phosphorylated tau interact abnormally with DNM1L, resulting in mitochondrial dysfunction and hastening AD progression [190,191,192,193,194,195,196,197,198]. Due to the integral role that DNM1L plays in normal neuronal function in the relative absence of Aβ and phosphorylated tau [199], ATRA may indirectly beneficially modulate DNM1L activity by α-secretase-mediated prevention of Aβ accumulation. Further investigation into the transcriptional control of DNM1L, the causes of its dysregulation, and its distinct roles in neuroprotection or neurotoxicity in AD and non-AD brains is needed before drawing definitive conclusions [200].

2.11. The Upregulation of Histone Deacetylases (HDACs) Suggests Epigenetic Changes That Interfere with RAR-and Nrf2-Mediated Transcription

Rates of transcription of ATRA-dependent genes involve mechanistic, epigenetic, and co-repressor factors. The inconsistency between the upregulation of Nrf2 mRNA expression and the downregulation of Nrf2-dependent gene targets (Table 3) led us to suspect that epigenetic mechanisms could play a role. Histone deacetylases (HDACs), together with histone acetyltransferases (HATs), set acetylation/deacetylation balance, allowing tight control over chromatin structure and gene transcription [201]. Among Class I HDACs, we found the most significant dysregulation in HDAC1 transcript from post-mortem AD hippocampus (Table 4). To a lesser extent, Class II HDACs, HDAC4 and HDAC7, were significantly upregulated. Together, we observed AD-related increases in HDAC expression, suggesting that relatively global epigenetic silencing of gene transcription underlies AD-related learning impairments. Therefore, even if ATRA were to be locally present at sufficient levels, histone deacetylation may preserve the closed chromatin conformation and promoter inaccessibility that downregulate critical ATRA- and Nrf2-induced gene expression. The finding of HDAC upregulation across class I/II HDACs suggests that a pan-HDAC inhibitor may counteract epigenetic changes, offering hope in treating AD onset and progression [202,203]. Indeed, HDAC inhibition has shown promising effects in AD mouse models [203,204].

2.12. Age-Related Epigenetic Changes Associated with RARE, PPARE, and ARE

Histones 3 and 4 (H3 and H4, respectively) have been implicated in the regulation of Nrf2 gene expression. H4 acetylation has been shown to increase transcription of Nrf2-dependent genes [205], while H3 and H4 deacetylation have been shown to reduce Nrf2 gene expression in microglia [206]. H3 acetylation is associated with unmethylated CpG islands in the Nrf2 promoter [207]. These results suggest that a HDAC inhibitor may optimally restore repressed Nrf2 expression. However, this therapeutic intervention only accounts for Nrf2 gene silencing attributed to epigenetic changes and does not account for Nrf2 inhibition or downregulation attributed to dysregulation of Nrf2 at the protein-protein interaction or transcription factor levels of regulation, such as by Keap1, GSK-3β, Bach1, p53, Hrd1, and miRNAs [208,209].
In addition to the decline in hippocampal ATRA levels due to age-related homeostatic collapse of the liver-brain axis [57], AD-related epigenetic changes may directly interfere with RAR- and PPAR-mediated transcriptional activity. In post-mortem AD brains and mouse models, histone deacetylation has been heavily implicated in the progression of AD [210,211,212,213,214,215]. Furthermore, VA deficient mice have lower levels of histone acetylation due to dysregulation of CBP-dependent mechanisms [216]. The therapeutic employment of class I/II HDAC inhibitors has shown wide-spread promise in treating AD by improving learning, memory, and behavioral functions, reducing neuroinflammation, and decreasing levels of Aβ and phosphorylated tau [203,204,217,218,219,220,221,222,223,224,225,226,227].
On the basis of these observations, we propose the following working model. Under conditions of ATRA deficiency, the unliganded RAR/RXR heterodimer binds to HDAC3 via co-repressors nuclear receptor corepressor (NCOR) or Silencing Mediator of Retinoid and Thyroid Hormone Receptors (SMRT) as well as corepressor CDK2-associated cullin domain 1 (CACUL1) [228,229,230]. HDAC3 functions by deacetylating nearby histones, further ensuring transcriptional inactivity of ATRA- and PPAR-sensitive genes [231] (Figure 4A). In the presence of ATRA, the liganded RAR/RXR heterodimer complex binds to histone acetyltransferases (HATs) such as CREB binding protein/p300 (CBP/p300), steroid receptor coactivator/p160 (SRC/p160), and p300/CBP-associated factor (P/CAF) (Figure 5A). Acetylation of histones by HATs promotes an open chromatin structure, increasing transcription. Histone methyltransferases (HMTs) such as coactivator associated arginine methyltransferase 1 (CARM1) and protein arginine methyltransferase 1 (PRMT1) are also recruited, responsible for methylating arginine residues on histones that can further activate the complex and increase gene expression [232], although histone methylation can also be silencing, with the effects depending on context and methylation pattern [233]. The association of PRMT1 with a RAR/RXR heterodimer bound to a RARE allows for a localized open chromatin conformation and subsequent transcription of ATRA-sensitive genes (Figure 5A).
ATRA depletion is also predicted to negatively affect PPARβ/δ-mediated gene expression. Unliganded RARα recruits NCOR or SMRT, decreasing ATRA-PPARβ/δ gene transcription [234] (Figure 4A). Similarly to RARα, these co-repressors are removed with the binding of ATRA to PPARβ/δ. Subsequent heterodimerization with RXR recruits numerous co-activators: peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α), switch/sucrose non-fermentable (SWI/SNF), CBP/p300, and steroid receptor coactivator 1 (SRC-1), the latter two of which are HATs [235,236]. The recruitment of these transcriptional co-activators and histone acetyltransferases increases ATRA-PPARβ/δ-RXR-sensitive gene expression, ultimately enhancing neuroprotection against inflammation and OS (Figure 5A).
Finally, when ATRA is present and bound to RARα, the complex binds to the Neh7 region on Nrf2, sequestering bioavailable Nrf2 and functioning as an ARE-bound co-repressor. We hypothesize that ATRA depletion, as well as depletion of other antioxidants, causes an increase in OS due to the lack of exogenous AO effect, resulting in the activation of compensatory Nrf2-mediated endogenous AO defenses (Figure 3B). However, histone deacetylation may progressively reduce Nrf2-sensitive gene expression, despite elevated OS. We propose a model in which the degree to which ATRA-RARα-Nrf2 sequestration occurs is contingent on cellular concentrations of ATRA. Under this model, excessive levels of ATRA lead to Nrf2 sequestration, but moderate ATRA levels do not, allowing Nrf2 to exert its homeostatic effects by recruiting c-Jun, JunD, activating transcription factor 4 (ATF4), and the HAT CBP/p300. It has been shown that CBP/p300 not only acetylates nearby histones to promote an open chromatin conformation, but also directly acetylates Nrf2 to augment the formation of the transcriptional complex [237]. By recruiting these transcription factors, there is increased gene expression of endogenous AOs such as NQO1/2, HO-1, GSTM4, and SOD1, providing neuroprotection that counteracts OS and attenuates the neurodegenerative process of AD [238] (Figure 5D).

3. Limitations, Open Questions, and Future Directions

Although we cover a variety of mechanistic aspects, there are a number of limitations associated with our assessment of human hippocampal transcriptomics and open questions remaining.
First, as we solely focused on the human dataset originally created by van Rooij and colleagues [107], we have not incorporated additional human datasets to compare and validate specific genes and RAR/PPARδ/β signaling pathways. Moreover, this data represents a snapshot in time during which AD has significantly progressed (Braak stage 5.5). The examination of the human hippocampal transcriptome earlier in AD pathogenesis (i.e., in mild cognitive impairment and/or earlier Braak stages) in the future will provide additional insights on the temporal relationship of VA status with respect to AD pathogenesis and progression. Nevertheless, the van Rooij et al. human dataset was adequately powered, statistically sound, and was highly heterogeneous, including both sexes and a mixture of APOE4 phenotypes. Therefore, observing these robust effects for the genes examined, even in the face of considerable heterogeneity, suggests that the involvement of VA mechanisms and ATRA-sensitive genes could be generalizable to additional time points and datasets.
Second, transcriptomics data is invaluable in providing data on thousands of mRNA transcripts; however, it is important to remember that gene expression may not perfectly correlate with protein expression. We assume that the binding of RARα/RXR to RAREs increases gene transcription at RARE sites on the promoter region of any given ATRA-sensitive gene (Figure 2), but this is most likely an oversimplification, as this is not always the case for any given ATRA-sensitive gene. We are acutely aware that additional bioinformatics resources exist on ATRA-sensitive gene expression. Several databases, such as TF2DNA and Enrichr, can provide detailed information on potential RAREs/PPAREs associated with specific genes. This work is limited in that it does not incorporate all of the available resources on ATRA-sensitive gene expression. A brief foray into Enrichr for any given ATRA-sensitive gene reveals a seemingly overwhelming number of additional transcription factors that may or may not be active with RARα-mediated gene transcription at RAREs, implying a complexity that is not yet well understood. Moreover, this study is based solely on RNA-seq data, which lacks cell type-specific information. We suspect that downregulation of ATRA-dependent mRNA transcripts is likely to occur in neuronal populations, whereas upregulation of ATRA-dependent mRNA transcripts could involve non-neuronal populations that relate to neuroinflammatory signaling mechanisms. Therefore, much work remains in determining how ATRA-sensitive gene expression in specific neuronal cell types is impacted by global ATRA deficiency that may be present in AD pathology.
Third, it is important to remember that VA plays important roles not only in the brain but also in other organ systems. A landmark rodent study demonstrating that age-related VA dysregulation involves disruption of the liver-brain axis [57] is a reminder that our understanding of the liver-brain axis in humans is at a nascent stage. The functional roles of retinoids in the brain could be impaired by insufficient dietary intake of VA, defective hepatic storage of VA, impaired hepatic release of retinol and RBP4, impaired transport of retinol-RBP4-TTR from liver to brain, and/or defective processing of retinol into ATRA in the human hippocampus [98]. In this study, we showed that there is a highly significant downregulation of hippocampal RBP4 (Table 1), which suggests impaired vascular transport of retinol from the liver to distant tissues, including the brain, assuming that hippocampal RBP4 downregulation is reflective of a similar downregulation in the liver. Although further study is needed to explore this possibility; such a finding is consistent with hepatic trapping of VA and dysregulation of the brain-liver axis as a potential upstream mechanism of ATRA deficiency in AD hippocampus. Therefore, it is likely that VA deficiency in brain is preceded by liver damage and hepatic trapping of VA stores in liver. Malabsorption of VA in gut, as well as dysbiosis, could contribute to early stages of disease pathogenesis, inducing local ATRA deficiency and inflammatory signaling. There is no doubt that co-morbidities occurring in midlife may increase the risk of AD-related dementia later in life. Therefore, a greater understanding of VA dysregulation in other disease processes (i.e., non-alcoholic fatty liver disease, cardiovascular dysfunction, obesity, diabetes) may provide important key insights into AD pathogenesis. These areas of research are exciting future directions but fall beyond the scope of this review.
Fourth, ATRA has many pleiotropic functions in the CNS. Not only is it considered an endogenous antioxidant with free radical scavenging capabilities like retinol and carotenoids [140,141,142,239], it is also a receptor ligand for RARs and PPARδ/β [31,139,240]. However, in addition to acting as hormone-like transcription factors at the genomic level, RARs also are known to exhibit non-genomic actions [79,241,242]. At a synaptic level, ATRA balances excitatory and inhibitory synaptic strength as part of its role in homeostatic plasticity and in preventing runaway Hebbian synaptic plasticity [242]. ATRA has also been shown to contribute to the cellular regulation of kinases and protein translation [164]. Moreover, ATRA synthesis appears to be tightly controlled by intracellular calcium dynamics [243]. Finally, ATRA plays an important role in neurogenesis, but these actions may be, in part, mediated by RAR-independent, non-genomic actions of ATRA [244], which have implications for neurodegenerative diseases [245]. These non-genomic mechanisms of ATRA are interesting and relevant to Alzheimer’s disease pathogenesis. We have limited this review to focus on the genomic actions of ATRA. However, it is likely that ATRA deficiency in AD would alter non-genomic aspects of ATRA signaling. These are interesting directions that can be examined in the future.
Fifth, although our foray through ATRA-sensitive genes and RAR co-repressors make a good case for ATRA deficiency, not all ATRA-sensitive genes exhibit downregulation consistent with ATRA deficiency. To this point, our initial examination of ATRA-sensitive transcripts does not directly support changes in the balance between amyloidogenic vs. non-amyloidogenic pathways in human AD. In this human dataset, based on previous studies (see above), we had predicted that the most critical of ATRA-sensitive genes to be the α-secretase ADAM10. Under amyloidogenic conditions, we predicted that ADAM10 mRNA would be downregulated relative to BACE1 mRNA, setting the stage for amyloidogenic pathway activation, accounting for AD progression. Surprisingly, we found ADAM10 mRNA to be significantly upregulated relative to BACE1 in this AD dataset (Table 5). As there is a known RARE within the ADAM10 promoter, this observation is inconsistent with a simple interpretation of ATRA deficiency. However, both ADAM10 and BACE1 had fairly weak DE scores (0.03 and 0.01, respectively), raising the question as to whether additional ADAM family members and/or BACE genes may contribute (Table 5). Given the constraint that we are primarily examining late-stage AD, additional compensatory mechanisms may be at play that complicate the interpretation of ADAM10 mRNA expression. We suspect that alternative transcriptional activators of ADAM10, such as PPARα, and especially SIRT1 [29,47,246], may partly account for this discrepancy, since both transcripts are significantly upregulated in this AD dataset (Table 5). Additional proteolytic ADAM family metalloproteinases are detected in this dataset (i.e., ADAM8, ADAM17, ADAM33), but these are also significantly upregulated similarly to ADAM10. It is possible that there are additional unknown compensatory mechanisms that come into play in an attempt to restore homeostasis. Finally, transcriptional upregulation may be a compensatory mechanism and may not necessarily translate to protein expression, giving no insight into potential downstream mechanisms. Further analysis, using additional datasets, and especially earlier timepoints, is necessary to interpret the broader meaning of this finding. That this simple prediction fails suggests that intrinsic transcriptional mechanisms are far more complex and will take more time and effort to unravel the underlying molecular mechanisms.
Sixth, HDAC inhibitors are not the only useful agents in reversing age-related epigenetic changes. For example, sulforaphane, a DNA methyltrasferase (DNMT) inhibitor found in cruciferous vegetables, upregulates Nrf2 expression by decreasing the methylation of cytosine residues in promoter CpG dinucleotides, lifting the epigenetic suppression of Nrf2 as well as many other genes. This action was also found to decrease levels of Aβ, ROS, and neuroinflammation in mouse models [247]. Similarly, resveratrol downregulates the expression of DNMTs, which decreases methylation at the Nrf2 promoter site. This increases Nrf2 expression and decreases levels of OS [248]. Interestingly, resveratrol has also been shown to reverse ATRA resistance by demethylating the CRABP2 gene, responsible for nuclear import of ATRA and its subsequent transfer to RARα [249]. There is even some evidence that the herbal supplement curcumin and exercise may also contribute to the demethylation of CpG islands in the promoter site of the Nrf2 gene and increase transcriptional expression [250,251]. Therefore, the incorporation of epigenetic changes into our working model is far from complete, with future drug combinations between nuclear receptor ligands, HDAC inhibitors, and DNMT inhibitors worthwhile to explore in future studies.
Finally, SIRT1, a class III HDAC, has been proposed to be neuroprotective against AD by deacetylating CRABP2 and RARβ, resulting in increased expression of α-secretase and reducing amyloidogenesis [252,253,254]. Moreover, it has even been proposed that SIRT1 has a direct agonist effect on RARβ that activates transcription of α-secretase [255]. Despite a previous retraction [256], this area of research has been extremely active. However, because these mechanisms likely involve DNA hypermethylation [257,258,259,260,261,262,263,264], we feel that these areas are beyond the scope of this review and best saved for a future article dedicated to this focus.

4. Conclusions

This is the first review to explicitly discuss ATRA-sensitive genes within the context of a human hippocampal transcriptomics dataset. We have explored possible molecular mechanisms stemming from select dysregulated genes in a publicly available hippocampal transcriptomic dataset from postmortem AD and control brains [107]. First, we demonstrate that a number of genes mediating retinol transport, ATRA synthesis, and ATRA metabolism are dysregulated in hippocampal tissue from post-mortem AD brains, supporting the hypothesis that ATRA deficiency occurs in the human hippocampus in AD. Second, we discovered transcriptional upregulation of RAR-related co-repressor genes, corroborating the idea of transcriptional repression of RAR-mediated transcription. Third, we noted increased expression of OS and neuroinflammatory genes is consistent with ATRA depletion. Interestingly, we encountered evidence from the cancer field of crosstalk between Nrf2- and RAR-mediated signaling, supporting the idea that oxidative stress-mediated Nrf2 upregulation is consistent with depletion of VA and other lipophilic vitamins (i.e., E and K) thought to have ROS scavenging activity. Moreover, increased NFKB1 and NFKB2 transcripts are associated with neuroinflammation which may be caused in part by oxidative stress. Interestingly, we noted that many targets of Nrf2 were not upregulated (Table 3), suggesting an inability of Nrf2 to modulate transcription. These observations raised the possibility that HDAC upregulation (Table 4) may partly account for the failure of Nrf2 targets to respond to Nrf2 upregulation.
Based on these observations, we have developed a working model of transcriptional mechanisms in AD, centering on ATRA deficiency and age-dependent epigenetic silencing (Figure 5). We propose that VA sufficiency combined with HDAC inhibition could sustain cognition and potentially avoid AD. Interestingly, we found sparse evidence for downregulation of ADAM10 in AD, as expected from ATRA deficiency (Table 5). However, given the limitations stated above, potential compensation effects late in the disease process may explain this observation, as well as other ADAM family members that may complicate this interpretation. Alternatively, our findings suggest renewed appreciation in ATRA as a ROS scavenger and the role of antioxidant depletion, generally, in causing oxidative stress and triggering of antioxidant defenses. Nevertheless, we feel that disruption of the balance between non-amyloidogenic and amyloidogenic pathways is an essential component of AD and should still be considered as a working model of AD (Figure 1). Furthermore, since ATRA binds to PPARδ/β receptors with high affinity [31], targets of PPARδ/β signaling also would be dysregulated, further contributing to OS elevation, neuroinflammation, Aβ plaque formation, and phosphorylated tau, which together accelerate AD onset and progression [51]. Lastly, we predict that Nrf2-mediated endogenous AOs are indeed engaged early in AD to combat the rising ROS. However, increased expression of HDACs and aberrant epigenetic silencing may explain global downregulation of RAR- and Nrf2-dependent genes in AD.
Overall, our mechanistic insights point towards VA supplementation, combined with HDAC inhibition, as a plausible AD prevention strategy (Figure 5D). Although several HDAC inhibitors are approved for use as drugs (i.e., vorinostat), a growing number of naturally occurring active metabolites in food are appreciated as HDAC inhibitors [265,266], many of which are associated with Mediterranean diet and healthy aging. Therefore, it is possible that a particular combination of nutrients, including not only VA sufficiency but also HDAC inhibition across lifespan, promotes healthy aging, longevity, and AD prevention.

Author Contributions

Conceptualization, J.A. and J.J.L.; investigation, J.A. and J.J.L.; writing—original draft preparation, J.A.; writing—review and editing, A.H. and J.J.L.; visualization, J.A. and J.J.L.; supervision, J.J.L.; funding acquisition, J.A. and J.J.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIH R01 AG071859 and NIH R01 AG073826 (J.J.L.). J.A. was supported by a TTUHSC Medical Student Summer Research Program scholarship.

Data Availability Statement

All data discussed are available via Supplementary Materials by van Rooij and colleagues [107].

Acknowledgments

We thank Chhanda Bose, Sharda Singh, and Boris Decourt for helpful comments and Kevin Pruitt and Igor Ponomarev for comments on an earlier stage of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 2023 Alzheimer’s disease facts and figures. Alzheimers Dement. 2023, 19, 1598–1695. [CrossRef] [PubMed]
  2. Murray, C.J.; Atkinson, C.; Bhalla, K.; Birbeck, G.; Burstein, R.; Chou, D.; Dellavalle, R.; Danaei, G.; Ezzati, M.; Fahimi, A.; et al. The state of US health, 1990–2010: Burden of diseases, injuries, and risk factors. JAMA 2013, 310, 591–608. [Google Scholar] [CrossRef] [PubMed]
  3. Lim, S.S.; Vos, T.; Flaxman, A.D.; Danaei, G.; Shibuya, K.; Adair-Rohani, H.; Amann, M.; Anderson, H.R.; Andrews, K.G.; Aryee, M.; et al. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012, 380, 2224–2260. [Google Scholar] [CrossRef]
  4. Alford, S.; Patel, D.; Perakakis, N.; Mantzoros, C.S. Obesity as a risk factor for Alzheimer’s disease: Weighing the evidence. Obes. Rev. 2018, 19, 269–280. [Google Scholar] [CrossRef]
  5. Lamar, M.; Boots, E.A.; Arfanakis, K.; Barnes, L.L.; Schneider, J.A. Common Brain Structural Alterations Associated with Cardiovascular Disease Risk Factors and Alzheimer’s Dementia: Future Directions and Implications. Neuropsychol. Rev. 2020, 30, 546–557. [Google Scholar] [CrossRef]
  6. Zhang, J.; Chen, C.; Hua, S.; Liao, H.; Wang, M.; Xiong, Y.; Cao, F. An updated meta-analysis of cohort studies: Diabetes and risk of Alzheimer’s disease. Diabetes Res. Clin. Pract. 2017, 124, 41–47. [Google Scholar] [CrossRef]
  7. Whitmer, R.A.; Gustafson, D.R.; Barrett-Connor, E.; Haan, M.N.; Gunderson, E.P.; Yaffe, K. Central obesity and increased risk of dementia more than three decades later. Neurology 2008, 71, 1057–1064. [Google Scholar] [CrossRef]
  8. Chen, T.B.; Yiao, S.Y.; Sun, Y.; Lee, H.J.; Yang, S.C.; Chiu, M.J.; Chen, T.F.; Lin, K.N.; Tang, L.Y.; Lin, C.C.; et al. Comorbidity and dementia: A nationwide survey in Taiwan. PLoS ONE 2017, 12, e0175475. [Google Scholar] [CrossRef]
  9. Samadi, M.; Moradi, S.; Moradinazar, M.; Mostafai, R.; Pasdar, Y. Dietary pattern in relation to the risk of Alzheimer’s disease: A systematic review. Neurol. Sci. 2019, 40, 2031–2043. [Google Scholar] [CrossRef] [PubMed]
  10. Scarmeas, N.; Anastasiou, C.A.; Yannakoulia, M. Nutrition and prevention of cognitive impairment. Lancet Neurol. 2018, 17, 1006–1015. [Google Scholar] [CrossRef] [PubMed]
  11. Vassilaki, M.; Aakre, J.A.; Syrjanen, J.A.; Mielke, M.M.; Geda, Y.E.; Kremers, W.K.; Machulda, M.M.; Alhurani, R.E.; Staubo, S.C.; Knopman, D.S.; et al. Mediterranean Diet, Its Components, and Amyloid Imaging Biomarkers. J. Alzheimers Dis. 2018, 64, 281–290. [Google Scholar] [CrossRef]
  12. Armstrong, R.A. The molecular biology of senile plaques and neurofibrillary tangles in Alzheimer’s disease. Folia Neuropathol. 2009, 47, 289–299. [Google Scholar] [PubMed]
  13. Wang, X.; Wang, W.; Li, L.; Perry, G.; Lee, H.G.; Zhu, X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim. Biophys. Acta 2014, 1842, 1240–1247. [Google Scholar] [CrossRef] [PubMed]
  14. Luo, J.; Mills, K.; le Cessie, S.; Noordam, R.; van Heemst, D. Ageing, age-related diseases and oxidative stress: What to do next? Ageing Res. Rev. 2020, 57, 100982. [Google Scholar] [CrossRef]
  15. Kryscio, R.J.; Abner, E.L.; Caban-Holt, A.; Lovell, M.; Goodman, P.; Darke, A.K.; Yee, M.; Crowley, J.; Schmitt, F.A. Association of Antioxidant Supplement Use and Dementia in the Prevention of Alzheimer’s Disease by Vitamin E and Selenium Trial (PREADViSE). JAMA Neurol. 2017, 74, 567–573. [Google Scholar] [CrossRef]
  16. Davies, M.; Holt, G. Why antioxidant therapies have failed in clinical trials. J. Theor. Biol. 2018, 457, 1–5. [Google Scholar] [CrossRef] [PubMed]
  17. Quadro, L.; Blaner, W.S.; Salchow, D.J.; Vogel, S.; Piantedosi, R.; Gouras, P.; Freeman, S.; Cosma, M.P.; Colantuoni, V.; Gottesman, M.E. Impaired retinal function and vitamin A availability in mice lacking retinol-binding protein. EMBO J. 1999, 18, 4633–4644. [Google Scholar] [CrossRef]
  18. Episkopou, V.; Maeda, S.; Nishiguchi, S.; Shimada, K.; Gaitanaris, G.A.; Gottesman, M.E.; Robertson, E.J. Disruption of the transthyretin gene results in mice with depressed levels of plasma retinol and thyroid hormone. Proc. Natl. Acad. Sci. USA 1993, 90, 2375–2379. [Google Scholar] [CrossRef]
  19. Kelly, M.; Widjaja-Adhi, M.A.; Palczewski, G.; von Lintig, J. Transport of vitamin A across blood-tissue barriers is facilitated by STRA6. FASEB J. 2016, 30, 2985–2995. [Google Scholar] [CrossRef]
  20. Napoli, J.L. Cellular retinoid binding-proteins, CRBP, CRABP, FABP5: Effects on retinoid metabolism, function and related diseases. Pharmacol. Ther. 2017, 173, 19–33. [Google Scholar] [CrossRef]
  21. Michalik, L.; Wahli, W. Guiding ligands to nuclear receptors. Cell 2007, 129, 649–651. [Google Scholar] [CrossRef] [PubMed]
  22. Schug, T.T.; Berry, D.C.; Shaw, N.S.; Travis, S.N.; Noy, N. Opposing effects of retinoic acid on cell growth result from alternate activation of two different nuclear receptors. Cell 2007, 129, 723–733. [Google Scholar] [CrossRef]
  23. Zhou, N.; Yao, Y.; Ye, H.; Zhu, W.; Chen, L.; Mao, Y. Abscisic-acid-induced cellular apoptosis and differentiation in glioma via the retinoid acid signaling pathway. Int. J. Cancer 2016, 138, 1947–1958. [Google Scholar] [CrossRef] [PubMed]
  24. Campos, B.; Centner, F.S.; Bermejo, J.L.; Ali, R.; Dorsch, K.; Wan, F.; Felsberg, J.; Ahmadi, R.; Grabe, N.; Reifenberger, G.; et al. Aberrant expression of retinoic acid signaling molecules influences patient survival in astrocytic gliomas. Am. J. Pathol. 2011, 178, 1953–1964. [Google Scholar] [CrossRef]
  25. Aranda, A.; Pascual, A. Nuclear hormone receptors and gene expression. Physiol. Rev. 2001, 81, 1269–1304. [Google Scholar] [CrossRef]
  26. de Urquiza, A.M.; Liu, S.; Sjoberg, M.; Zetterstrom, R.H.; Griffiths, W.; Sjovall, J.; Perlmann, T. Docosahexaenoic acid, a ligand for the retinoid X receptor in mouse brain. Science 2000, 290, 2140–2144. [Google Scholar] [CrossRef] [PubMed]
  27. Tafti, M.; Ghyselinck, N.B. Functional implication of the vitamin A signaling pathway in the brain. Arch. Neurol. 2007, 64, 1706–1711. [Google Scholar] [CrossRef]
  28. Heyman, R.A.; Mangelsdorf, D.J.; Dyck, J.A.; Stein, R.B.; Eichele, G.; Evans, R.M.; Thaller, C. 9-cis retinoic acid is a high affinity ligand for the retinoid X receptor. Cell 1992, 68, 397–406. [Google Scholar] [CrossRef]
  29. Peron, R.; Vatanabe, I.P.; Manzine, P.R.; Camins, A.; Cominetti, M.R. Alpha-Secretase ADAM10 Regulation: Insights into Alzheimer’s Disease Treatment. Pharmaceuticals 2018, 11, 12. [Google Scholar] [CrossRef]
  30. Strosznajder, A.K.; Wojtowicz, S.; Jezyna, M.J.; Sun, G.Y.; Strosznajder, J.B. Recent Insights on the Role of PPAR-beta/delta in Neuroinflammation and Neurodegeneration, and Its Potential Target for Therapy. Neuromolecular Med. 2021, 23, 86–98. [Google Scholar] [CrossRef]
  31. Shaw, N.; Elholm, M.; Noy, N. Retinoic acid is a high affinity selective ligand for the peroxisome proliferator-activated receptor beta/delta. J. Biol. Chem. 2003, 278, 41589–41592. [Google Scholar] [CrossRef] [PubMed]
  32. Iwashita, A.; Muramatsu, Y.; Yamazaki, T.; Muramoto, M.; Kita, Y.; Yamazaki, S.; Mihara, K.; Moriguchi, A.; Matsuoka, N. Neuroprotective efficacy of the peroxisome proliferator-activated receptor delta-selective agonists in vitro and in vivo. J. Pharmacol. Exp. Ther. 2007, 320, 1087–1096. [Google Scholar] [CrossRef]
  33. Tong, Q.; Wu, L.; Gao, Q.; Ou, Z.; Zhu, D.; Zhang, Y. PPARbeta/delta Agonist Provides Neuroprotection by Suppression of IRE1alpha-Caspase-12-Mediated Endoplasmic Reticulum Stress Pathway in the Rotenone Rat Model of Parkinson’s Disease. Mol. Neurobiol. 2016, 53, 3822–3831. [Google Scholar] [CrossRef] [PubMed]
  34. Colas, J.; Chessel, N.; Ouared, A.; Gruz-Gibelli, E.; Marin, P.; Herrmann, F.R.; Savioz, A. Neuroprotection against Amyloid-beta-Induced DNA Double-Strand Breaks Is Mediated by Multiple Retinoic Acid-Dependent Pathways. Neural Plast. 2020, 2020, 9369815. [Google Scholar] [CrossRef]
  35. Barroso, E.; del Valle, J.; Porquet, D.; Vieira Santos, A.M.; Salvado, L.; Rodriguez-Rodriguez, R.; Gutierrez, P.; Anglada-Huguet, M.; Alberch, J.; Camins, A.; et al. Tau hyperphosphorylation and increased BACE1 and RAGE levels in the cortex of PPARbeta/delta-null mice. Biochim. Biophys. Acta 2013, 1832, 1241–1248. [Google Scholar] [CrossRef]
  36. Altinoz, M.A.; Ozpinar, A. PPAR-delta and erucic acid in multiple sclerosis and Alzheimer’s Disease. Likely benefits in terms of immunity and metabolism. Int. Immunopharmacol. 2019, 69, 245–256. [Google Scholar] [CrossRef]
  37. Aleshin, S.; Reiser, G. Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) protects against ceramide-induced cellular toxicity in rat brain astrocytes and neurons by activation of ceramide kinase. Mol. Cell Neurosci. 2014, 59, 127–134. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, W.J.; Ham, S.A.; Lee, G.H.; Choi, M.J.; Yoo, H.; Paek, K.S.; Lim, D.S.; Hong, K.; Hwang, J.S.; Seo, H.G. Activation of peroxisome proliferator-activated receptor delta suppresses BACE1 expression by up-regulating SOCS1 in a JAK2/STAT1-dependent manner. J. Neurochem. 2019, 151, 370–385. [Google Scholar] [CrossRef] [PubMed]
  39. Malm, T.; Mariani, M.; Donovan, L.J.; Neilson, L.; Landreth, G.E. Activation of the nuclear receptor PPARdelta is neuroprotective in a transgenic mouse model of Alzheimer’s disease through inhibition of inflammation. J. Neuroinflamm. 2015, 12, 7. [Google Scholar] [CrossRef]
  40. An, Y.Q.; Zhang, C.T.; Du, Y.; Zhang, M.; Tang, S.S.; Hu, M.; Long, Y.; Sun, H.B.; Hong, H. PPARdelta agonist GW0742 ameliorates Abeta1-42-induced hippocampal neurotoxicity in mice. Metab. Brain Dis. 2016, 31, 663–671. [Google Scholar] [CrossRef]
  41. Kalinin, S.; Richardson, J.C.; Feinstein, D.L. A PPARdelta agonist reduces amyloid burden and brain inflammation in a transgenic mouse model of Alzheimer’s disease. Curr. Alzheimer Res. 2009, 6, 431–437. [Google Scholar] [CrossRef]
  42. Tong, M.; Dominguez, C.; Didsbury, J.; de la Monte, S.M. Targeting Alzheimer’s Disease Neuro-Metabolic Dysfunction with a Small Molecule Nuclear Receptor Agonist (T3D-959) Reverses Disease Pathologies. J. Alzheimers Dis. Parkinsonism 2016, 6, 238. [Google Scholar] [CrossRef]
  43. Tong, M.; Deochand, C.; Didsbury, J.; de la Monte, S.M. T3D-959: A Multi-Faceted Disease Remedial Drug Candidate for the Treatment of Alzheimer’s Disease. J. Alzheimers Dis. 2016, 51, 123–138. [Google Scholar] [CrossRef] [PubMed]
  44. Chamberlain, S.; Gabriel, H.; Strittmatter, W.; Didsbury, J. An Exploratory Phase IIa Study of the PPAR delta/gamma Agonist T3D-959 Assessing Metabolic and Cognitive Function in Subjects with Mild to Moderate Alzheimer’s Disease. J. Alzheimers Dis. 2020, 73, 1085–1103. [Google Scholar] [CrossRef] [PubMed]
  45. Luo, R.; Su, L.Y.; Li, G.; Yang, J.; Liu, Q.; Yang, L.X.; Zhang, D.F.; Zhou, H.; Xu, M.; Fan, Y.; et al. Activation of PPARA-mediated autophagy reduces Alzheimer disease-like pathology and cognitive decline in a murine model. Autophagy 2020, 16, 52–69. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, M.; Qian, C.; Zheng, Z.G.; Qian, F.; Wang, Y.; Thu, P.M.; Zhang, X.; Zhou, Y.; Tu, L.; Liu, Q.; et al. Jujuboside A promotes Abeta clearance and ameliorates cognitive deficiency in Alzheimer’s disease through activating Axl/HSP90/PPARgamma pathway. Theranostics 2018, 8, 4262–4278. [Google Scholar] [CrossRef] [PubMed]
  47. Corbett, G.T.; Gonzalez, F.J.; Pahan, K. Activation of peroxisome proliferator-activated receptor alpha stimulates ADAM10-mediated proteolysis of APP. Proc. Natl. Acad. Sci. USA 2015, 112, 8445–8450. [Google Scholar] [CrossRef]
  48. Yuan, X.Z.; Sun, S.; Tan, C.C.; Yu, J.T.; Tan, L. The Role of ADAM10 in Alzheimer’s Disease. J. Alzheimers Dis. 2017, 58, 303–322. [Google Scholar] [CrossRef]
  49. Wojtowicz, S.; Strosznajder, A.K.; Jezyna, M.; Strosznajder, J.B. The Novel Role of PPAR Alpha in the Brain: Promising Target in Therapy of Alzheimer’s Disease and Other Neurodegenerative Disorders. Neurochem. Res. 2020, 45, 972–988. [Google Scholar] [CrossRef]
  50. Mandrekar-Colucci, S.; Landreth, G.E. Nuclear receptors as therapeutic targets for Alzheimer’s disease. Expert. Opin. Ther. Targets 2011, 15, 1085–1097. [Google Scholar] [CrossRef]
  51. de la Monte, S.M.; Wands, J.R. Molecular indices of oxidative stress and mitochondrial dysfunction occur early and often progress with severity of Alzheimer’s disease. J. Alzheimers Dis. 2006, 9, 167–181. [Google Scholar] [CrossRef]
  52. Allenby, G.; Bocquel, M.T.; Saunders, M.; Kazmer, S.; Speck, J.; Rosenberger, M.; Lovey, A.; Kastner, P.; Grippo, J.F.; Chambon, P.; et al. Retinoic acid receptors and retinoid X receptors: Interactions with endogenous retinoic acids. Proc. Natl. Acad. Sci. USA 1993, 90, 30–34. [Google Scholar] [CrossRef]
  53. Tate, B.F.; Levin, A.A.; Grippo, J.F. The discovery of 9-cis retinoic acid: A hormone that binds the retinoid-X receptor. Trends Endocrinol. Metab. 1994, 5, 189–194. [Google Scholar] [CrossRef]
  54. Yu, S.; Levi, L.; Siegel, R.; Noy, N. Retinoic acid induces neurogenesis by activating both retinoic acid receptors (RARs) and peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta). J. Biol. Chem. 2012, 287, 42195–42205. [Google Scholar] [CrossRef]
  55. Holzapfel, J.; Heun, R.; Lutjohann, D.; Jessen, F.; Maier, W.; Kolsch, H. PPARD haplotype influences cholesterol metabolism but is no risk factor of Alzheimer’s disease. Neurosci. Lett. 2006, 408, 57–61. [Google Scholar] [CrossRef]
  56. Helisalmi, S.; Vepsalainen, S.; Hiltunen, M.; Koivisto, A.M.; Salminen, A.; Laakso, M.; Soininen, H. Genetic study between SIRT1, PPARD, PGC-1alpha genes and Alzheimer’s disease. J. Neurol. 2008, 255, 668–673. [Google Scholar] [CrossRef] [PubMed]
  57. Dumetz, F.; Bure, C.; Alfos, S.; Bonneu, M.; Richard, E.; Touyarot, K.; Marie, A.; Schmitter, J.M.; Bosch-Bouju, C.; Pallet, V. Normalization of hippocampal retinoic acid level corrects age-related memory deficits in rats. Neurobiol. Aging 2020, 85, 1–10. [Google Scholar] [CrossRef] [PubMed]
  58. Cocco, S.; Diaz, G.; Stancampiano, R.; Diana, A.; Carta, M.; Curreli, R.; Sarais, L.; Fadda, F. Vitamin A deficiency produces spatial learning and memory impairment in rats. Neuroscience 2002, 115, 475–482. [Google Scholar] [CrossRef] [PubMed]
  59. Mingaud, F.; Mormede, C.; Etchamendy, N.; Mons, N.; Niedergang, B.; Wietrzych, M.; Pallet, V.; Jaffard, R.; Krezel, W.; Higueret, P.; et al. Retinoid hyposignaling contributes to aging-related decline in hippocampal function in short-term/working memory organization and long-term declarative memory encoding in mice. J. Neurosci. 2008, 28, 279–291. [Google Scholar] [CrossRef]
  60. Etchamendy, N.; Enderlin, V.; Marighetto, A.; Pallet, V.; Higueret, P.; Jaffard, R. Vitamin A deficiency and relational memory deficit in adult mice: Relationships with changes in brain retinoid signalling. Behav. Brain Res. 2003, 145, 37–49. [Google Scholar] [CrossRef] [PubMed]
  61. Wang, C.; Kane, M.A.; Napoli, J.L. Multiple retinol and retinal dehydrogenases catalyze all-trans-retinoic acid biosynthesis in astrocytes. J. Biol. Chem. 2011, 286, 6542–6553. [Google Scholar] [CrossRef]
  62. Jacobs, S.; Lie, D.C.; DeCicco, K.L.; Shi, Y.; DeLuca, L.M.; Gage, F.H.; Evans, R.M. Retinoic acid is required early during adult neurogenesis in the dentate gyrus. Proc. Natl. Acad. Sci. USA 2006, 103, 3902–3907. [Google Scholar] [CrossRef]
  63. Kojro, E.; Fahrenholz, F. The non-amyloidogenic pathway: Structure and function of alpha-secretases. Subcell. Biochem. 2005, 38, 105–127. [Google Scholar] [CrossRef]
  64. Corcoran, J.P.; So, P.L.; Maden, M. Disruption of the retinoid signalling pathway causes a deposition of amyloid beta in the adult rat brain. Eur. J. Neurosci. 2004, 20, 896–902. [Google Scholar] [CrossRef]
  65. Fahrenholz, F. The close link between retinoid signalling and the alpha-secretase ADAM10 and its potential for treating Alzheimer’s disease (commentary on Jarvis et al.). Eur. J. Neurosci. 2010, 32, 1245. [Google Scholar] [CrossRef]
  66. Jarvis, C.I.; Goncalves, M.B.; Clarke, E.; Dogruel, M.; Kalindjian, S.B.; Thomas, S.A.; Maden, M.; Corcoran, J.P. Retinoic acid receptor-alpha signalling antagonizes both intracellular and extracellular amyloid-beta production and prevents neuronal cell death caused by amyloid-beta. Eur. J. Neurosci. 2010, 32, 1246–1255. [Google Scholar] [CrossRef]
  67. Tippmann, F.; Hundt, J.; Schneider, A.; Endres, K.; Fahrenholz, F. Up-regulation of the alpha-secretase ADAM10 by retinoic acid receptors and acitretin. FASEB J. 2009, 23, 1643–1654. [Google Scholar] [CrossRef]
  68. Ding, Y.; Qiao, A.; Wang, Z.; Goodwin, J.S.; Lee, E.S.; Block, M.L.; Allsbrook, M.; McDonald, M.P.; Fan, G.H. Retinoic acid attenuates beta-amyloid deposition and rescues memory deficits in an Alzheimer’s disease transgenic mouse model. J. Neurosci. 2008, 28, 11622–11634. [Google Scholar] [CrossRef]
  69. Etchamendy, N.; Enderlin, V.; Marighetto, A.; Vouimba, R.M.; Pallet, V.; Jaffard, R.; Higueret, P. Alleviation of a selective age-related relational memory deficit in mice by pharmacologically induced normalization of brain retinoid signaling. J. Neurosci. 2001, 21, 6423–6429. [Google Scholar] [CrossRef]
  70. Gieng, S.H.; Green, M.H.; Green, J.B.; Rosales, F.J. Model-based compartmental analysis indicates a reduced mobilization of hepatic vitamin A during inflammation in rats. J. Lipid Res. 2007, 48, 904–913. [Google Scholar] [CrossRef]
  71. Hellmann-Regen, J.; Kronenberg, G.; Uhlemann, R.; Freyer, D.; Endres, M.; Gertz, K. Accelerated degradation of retinoic acid by activated microglia. J. Neuroimmunol. 2013, 256, 1–6. [Google Scholar] [CrossRef]
  72. Mandrekar-Colucci, S.; Landreth, G.E. Microglia and inflammation in Alzheimer’s disease. CNS Neurol. Disord. Drug Targets 2010, 9, 156–167. [Google Scholar] [CrossRef]
  73. Zhang, X.; Subbanna, S.; Williams, C.R.O.; Canals-Baker, S.; Smiley, J.F.; Wilson, D.A.; Das, B.C.; Saito, M. Anti-inflammatory Action of BT75, a Novel RARalpha Agonist, in Cultured Microglia and in an Experimental Mouse Model of Alzheimer’s Disease. Neurochem. Res. 2023, 48, 1958–1970. [Google Scholar] [CrossRef]
  74. Li, J.; Park, E.; Zhong, L.R.; Chen, L. Homeostatic synaptic plasticity as a metaplasticity mechanism—A molecular and cellular perspective. Curr. Opin. Neurobiol. 2019, 54, 44–53. [Google Scholar] [CrossRef]
  75. Walters, B.J.; Josselyn, S.A. Retinoic acid receptor plays both sides of homeostatic plasticity. Proc. Natl. Acad. Sci. USA 2019, 116, 6528–6530. [Google Scholar] [CrossRef]
  76. Groth, R.D.; Tsien, R.W. A role for retinoic acid in homeostatic plasticity. Neuron 2008, 60, 192–194. [Google Scholar] [CrossRef]
  77. Aoto, J.; Nam, C.I.; Poon, M.M.; Ting, P.; Chen, L. Synaptic signaling by all-trans retinoic acid in homeostatic synaptic plasticity. Neuron 2008, 60, 308–320. [Google Scholar] [CrossRef]
  78. Hsu, Y.T.; Li, J.; Wu, D.; Sudhof, T.C.; Chen, L. Synaptic retinoic acid receptor signaling mediates mTOR-dependent metaplasticity that controls hippocampal learning. Proc. Natl. Acad. Sci. USA 2019, 116, 7113–7122. [Google Scholar] [CrossRef]
  79. Woloszynowska-Fraser, M.U.; Kouchmeshky, A.; McCaffery, P. Vitamin A and Retinoic Acid in Cognition and Cognitive Disease. Annu. Rev. Nutr. 2020, 40, 247–272. [Google Scholar] [CrossRef]
  80. Berti, V.; Murray, J.; Davies, M.; Spector, N.; Tsui, W.H.; Li, Y.; Williams, S.; Pirraglia, E.; Vallabhajosula, S.; McHugh, P.; et al. Nutrient patterns and brain biomarkers of Alzheimer’s disease in cognitively normal individuals. J. Nutr. Health Aging 2015, 19, 413–423. [Google Scholar] [CrossRef]
  81. Jama, J.W.; Launer, L.J.; Witteman, J.C.; den Breeijen, J.H.; Breteler, M.M.; Grobbee, D.E.; Hofman, A. Dietary antioxidants and cognitive function in a population-based sample of older persons. The Rotterdam Study. Am. J. Epidemiol. 1996, 144, 275–280. [Google Scholar] [CrossRef]
  82. Zhao, X.; Yuan, L.; Feng, L.; Xi, Y.; Yu, H.; Ma, W.; Zhang, D.; Xiao, R. Association of dietary intake and lifestyle pattern with mild cognitive impairment in the elderly. J. Nutr. Health Aging 2015, 19, 164–168. [Google Scholar] [CrossRef]
  83. Zaman, Z.; Roche, S.; Fielden, P.; Frost, P.G.; Niriella, D.C.; Cayley, A.C. Plasma concentrations of vitamins A and E and carotenoids in Alzheimer’s disease. Age Ageing 1992, 21, 91–94. [Google Scholar] [CrossRef]
  84. Boccardi, V.; Arosio, B.; Cari, L.; Bastiani, P.; Scamosci, M.; Casati, M.; Ferri, E.; Bertagnoli, L.; Ciccone, S.; Rossi, P.D.; et al. Beta-carotene, telomerase activity and Alzheimer’s disease in old age subjects. Eur. J. Nutr. 2020, 59, 119–126. [Google Scholar] [CrossRef]
  85. Zeng, J.; Li, T.; Gong, M.; Jiang, W.; Yang, T.; Chen, J.; Liu, Y.; Chen, L. Marginal Vitamin A Deficiency Exacerbates Memory Deficits Following Abeta1-42 Injection in Rats. Curr. Alzheimer Res. 2017, 14, 562–570. [Google Scholar] [CrossRef]
  86. Rinaldi, P.; Polidori, M.C.; Metastasio, A.; Mariani, E.; Mattioli, P.; Cherubini, A.; Catani, M.; Cecchetti, R.; Senin, U.; Mecocci, P. Plasma antioxidants are similarly depleted in mild cognitive impairment and in Alzheimer’s disease. Neurobiol. Aging 2003, 24, 915–919. [Google Scholar] [CrossRef]
  87. Johnson, E.J.; Vishwanathan, R.; Johnson, M.A.; Hausman, D.B.; Davey, A.; Scott, T.M.; Green, R.C.; Miller, L.S.; Gearing, M.; Woodard, J.; et al. Relationship between Serum and Brain Carotenoids, alpha-Tocopherol, and Retinol Concentrations and Cognitive Performance in the Oldest Old from the Georgia Centenarian Study. J. Aging Res. 2013, 2013, 951786. [Google Scholar] [CrossRef] [PubMed]
  88. Huang, X.; Zhang, H.; Zhen, J.; Dong, S.; Guo, Y.; Van Halm-Lutterodt, N.; Yuan, L. Diminished circulating retinol and elevated alpha-TOH/retinol ratio predict an increased risk of cognitive decline in aging Chinese adults, especially in subjects with ApoE2 or ApoE4 genotype. Aging 2018, 10, 4066–4083. [Google Scholar] [CrossRef]
  89. Mullan, K.; Cardwell, C.R.; McGuinness, B.; Woodside, J.V.; McKay, G.J. Plasma Antioxidant Status in Patients with Alzheimer’s Disease and Cognitively Intact Elderly: A Meta-Analysis of Case-Control Studies. J. Alzheimers Dis. 2018, 62, 305–317. [Google Scholar] [CrossRef]
  90. Shahar, S.; Lee, L.K.; Rajab, N.; Lim, C.L.; Harun, N.A.; Noh, M.F.; Mian-Then, S.; Jamal, R. Association between vitamin A, vitamin E and apolipoprotein E status with mild cognitive impairment among elderly people in low-cost residential areas. Nutr. Neurosci. 2013, 16, 6–12. [Google Scholar] [CrossRef]
  91. Lopes da Silva, S.; Vellas, B.; Elemans, S.; Luchsinger, J.; Kamphuis, P.; Yaffe, K.; Sijben, J.; Groenendijk, M.; Stijnen, T. Plasma nutrient status of patients with Alzheimer’s disease: Systematic review and meta-analysis. Alzheimers Dement. 2014, 10, 485–502. [Google Scholar] [CrossRef] [PubMed]
  92. Zwilling, C.E.; Talukdar, T.; Zamroziewicz, M.K.; Barbey, A.K. Nutrient biomarker patterns, cognitive function, and fMRI measures of network efficiency in the aging brain. Neuroimage 2019, 188, 239–251. [Google Scholar] [CrossRef] [PubMed]
  93. Beydoun, M.A.; Canas, J.A.; Fanelli-Kuczmarski, M.T.; Maldonado, A.I.; Shaked, D.; Kivimaki, M.; Evans, M.K.; Zonderman, A.B. Association of Antioxidant Vitamins A, C, E and Carotenoids with Cognitive Performance over Time: A Cohort Study of Middle-Aged Adults. Nutrients 2020, 12, 3558. [Google Scholar] [CrossRef] [PubMed]
  94. Jimenez-Jimenez, F.J.; Molina, J.A.; de Bustos, F.; Orti-Pareja, M.; Benito-Leon, J.; Tallon-Barranco, A.; Gasalla, T.; Porta, J.; Arenas, J. Serum levels of beta-carotene, alpha-carotene and vitamin A in patients with Alzheimer’s disease. Eur. J. Neurol. 1999, 6, 495–497. [Google Scholar] [CrossRef] [PubMed]
  95. Bourdel-Marchasson, I.; Delmas-Beauvieux, M.C.; Peuchant, E.; Richard-Harston, S.; Decamps, A.; Reignier, B.; Emeriau, J.P.; Rainfray, M. Antioxidant defences and oxidative stress markers in erythrocytes and plasma from normally nourished elderly Alzheimer patients. Age Ageing 2001, 30, 235–241. [Google Scholar] [CrossRef]
  96. Andersen, L.F.; Jacobs, D.R., Jr.; Gross, M.D.; Schreiner, P.J.; Dale Williams, O.; Lee, D.H. Longitudinal associations between body mass index and serum carotenoids: The CARDIA study. Br. J. Nutr. 2006, 95, 358–365. [Google Scholar] [CrossRef]
  97. Min, K.B.; Min, J.Y. Association between leukocyte telomere length and serum carotenoid in US adults. Eur. J. Nutr. 2017, 56, 1045–1052. [Google Scholar] [CrossRef]
  98. Goodman, A.B.; Pardee, A.B. Evidence for defective retinoid transport and function in late onset Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2003, 100, 2901–2905. [Google Scholar] [CrossRef]
  99. Goodman, A.B. Retinoid receptors, transporters, and metabolizers as therapeutic targets in late onset Alzheimer disease. J. Cell Physiol. 2006, 209, 598–603. [Google Scholar] [CrossRef]
  100. Kolsch, H.; Lutjohann, D.; Jessen, F.; Popp, J.; Hentschel, F.; Kelemen, P.; Friedrichs, S.; Maier, T.A.; Heun, R. RXRA gene variations influence Alzheimer’s disease risk and cholesterol metabolism. J. Cell Mol. Med. 2009, 13, 589–598. [Google Scholar] [CrossRef]
  101. Vossel, K.A.; Beagle, A.J.; Rabinovici, G.D.; Shu, H.; Lee, S.E.; Naasan, G.; Hegde, M.; Cornes, S.B.; Henry, M.L.; Nelson, A.B.; et al. Seizures and epileptiform activity in the early stages of Alzheimer disease. JAMA Neurol. 2013, 70, 1158–1166. [Google Scholar] [CrossRef] [PubMed]
  102. Tao, H.; Zhao, J.; Zhou, X.; Ma, Z.; Chen, Y.; Sun, F.; Cui, L.; Zhou, H.; Cai, Y.; Chen, Y.; et al. Promoter Variants of the ADAM10 Gene and Their Roles in Temporal Lobe Epilepsy. Front. Neurol. 2016, 7, 108. [Google Scholar] [CrossRef]
  103. Zhu, X.; Li, X.; Zhu, M.; Xu, K.; Yang, L.; Han, B.; Huang, R.; Zhang, A.; Yao, H. Metalloprotease Adam10 suppresses epilepsy through repression of hippocampal neuroinflammation. J. Neuroinflamm. 2018, 15, 221. [Google Scholar] [CrossRef] [PubMed]
  104. Olsen, T.; Blomhoff, R. Retinol, Retinoic Acid, and Retinol-Binding Protein 4 are Differentially Associated with Cardiovascular Disease, Type 2 Diabetes, and Obesity: An Overview of Human Studies. Adv. Nutr. 2020, 11, 644–666. [Google Scholar] [CrossRef]
  105. Zhong, G.; Kirkwood, J.; Won, K.J.; Tjota, N.; Jeong, H.; Isoherranen, N. Characterization of Vitamin A Metabolome in Human Livers With and Without Nonalcoholic Fatty Liver Disease. J. Pharmacol. Exp. Ther. 2019, 370, 92–103. [Google Scholar] [CrossRef]
  106. Endres, K.; Fahrenholz, F.; Lotz, J.; Hiemke, C.; Teipel, S.; Lieb, K.; Tuscher, O.; Fellgiebel, A. Increased CSF APPs-alpha levels in patients with Alzheimer disease treated with acitretin. Neurology 2014, 83, 1930–1935. [Google Scholar] [CrossRef]
  107. van Rooij, J.G.J.; Meeter, L.H.H.; Melhem, S.; Nijholt, D.A.T.; Wong, T.H.; Netherlands Brain, B.; Rozemuller, A.; Uitterlinden, A.G.; van Meurs, J.G.; van Swieten, J.C. Hippocampal transcriptome profiling combined with protein-protein interaction analysis elucidates Alzheimer’s disease pathways and genes. Neurobiol. Aging 2019, 74, 225–233. [Google Scholar] [CrossRef]
  108. Balmer, J.E.; Blomhoff, R. Gene expression regulation by retinoic acid. J. Lipid Res. 2002, 43, 1773–1808. [Google Scholar] [CrossRef]
  109. Balmer, J.E.; Blomhoff, R. A robust characterization of retinoic acid response elements based on a comparison of sites in three species. J. Steroid Biochem. Mol. Biol. 2005, 96, 347–354. [Google Scholar] [CrossRef] [PubMed]
  110. Fang, M.; Li, Y.; Ren, J.; Hu, R.; Gao, X.; Chen, L. Epilepsy-Associated UBE3A Deficiency Downregulates Retinoic Acid Signalling Pathway. Front. Genet. 2021, 12, 681295. [Google Scholar] [CrossRef]
  111. Lalevee, S.; Anno, Y.N.; Chatagnon, A.; Samarut, E.; Poch, O.; Laudet, V.; Benoit, G.; Lecompte, O.; Rochette-Egly, C. Genome-wide in silico identification of new conserved and functional retinoic acid receptor response elements (direct repeats separated by 5 bp). J. Biol. Chem. 2011, 286, 33322–33334. [Google Scholar] [CrossRef]
  112. Niewiadomska-Cimicka, A.; Krzyzosiak, A.; Ye, T.; Podlesny-Drabiniok, A.; Dembele, D.; Dolle, P.; Krezel, W. Genome-wide Analysis of RARbeta Transcriptional Targets in Mouse Striatum Links Retinoic Acid Signaling with Huntington’s Disease and Other Neurodegenerative Disorders. Mol. Neurobiol. 2017, 54, 3859–3878. [Google Scholar] [CrossRef]
  113. Pujato, M.; Kieken, F.; Skiles, A.A.; Tapinos, N.; Fiser, A. Prediction of DNA binding motifs from 3D models of transcription factors; identifying TLX3 regulated genes. Nucleic Acids Res. 2014, 42, 13500–13512. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, E.Y.; Tan, C.M.; Kou, Y.; Duan, Q.; Wang, Z.; Meirelles, G.V.; Clark, N.R.; Ma’ayan, A. Enrichr: Interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinform. 2013, 14, 128. [Google Scholar] [CrossRef] [PubMed]
  115. Kuleshov, M.V.; Jones, M.R.; Rouillard, A.D.; Fernandez, N.F.; Duan, Q.; Wang, Z.; Koplev, S.; Jenkins, S.L.; Jagodnik, K.M.; Lachmann, A.; et al. Enrichr: A comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016, 44, W90–W97. [Google Scholar] [CrossRef] [PubMed]
  116. Xie, Z.; Bailey, A.; Kuleshov, M.V.; Clarke, D.J.B.; Evangelista, J.E.; Jenkins, S.L.; Lachmann, A.; Wojciechowicz, M.L.; Kropiwnicki, E.; Jagodnik, K.M.; et al. Gene Set Knowledge Discovery with Enrichr. Curr. Protoc. 2021, 1, e90. [Google Scholar] [CrossRef]
  117. Smith, W.C.; Nakshatri, H.; Leroy, P.; Rees, J.; Chambon, P. A retinoic acid response element is present in the mouse cellular retinol binding protein I (mCRBPI) promoter. EMBO J. 1991, 10, 2223–2230. [Google Scholar] [CrossRef]
  118. Husmann, M.; Hoffmann, B.; Stump, D.G.; Chytil, F.; Pfahl, M. A retinoic acid response element from the rat CRBPI promoter is activated by an RAR/RXR heterodimer. Biochem. Biophys. Res. Commun. 1992, 187, 1558–1564. [Google Scholar] [CrossRef]
  119. Loudig, O.; Babichuk, C.; White, J.; Abu-Abed, S.; Mueller, C.; Petkovich, M. Cytochrome P450RAI(CYP26) promoter: A distinct composite retinoic acid response element underlies the complex regulation of retinoic acid metabolism. Mol. Endocrinol. 2000, 14, 1483–1497. [Google Scholar] [CrossRef]
  120. Topletz, A.R.; Thatcher, J.E.; Zelter, A.; Lutz, J.D.; Tay, S.; Nelson, W.L.; Isoherranen, N. Comparison of the function and expression of CYP26A1 and CYP26B1, the two retinoic acid hydroxylases. Biochem. Pharmacol. 2012, 83, 149–163. [Google Scholar] [CrossRef]
  121. Idres, N.; Marill, J.; Flexor, M.A.; Chabot, G.G. Activation of retinoic acid receptor-dependent transcription by all-trans-retinoic acid metabolites and isomers. J. Biol. Chem. 2002, 277, 31491–31498. [Google Scholar] [CrossRef]
  122. Khatib, T.; Chisholm, D.R.; Whiting, A.; Platt, B.; McCaffery, P. Decay in Retinoic Acid Signaling in Varied Models of Alzheimer’s Disease and In-Vitro Test of Novel Retinoic Acid Receptor Ligands (RAR-Ms) to Regulate Protective Genes. J. Alzheimers Dis. 2020, 73, 935–954. [Google Scholar] [CrossRef] [PubMed]
  123. Leroy, P.; Nakshatri, H.; Chambon, P. Mouse retinoic acid receptor alpha 2 isoform is transcribed from a promoter that contains a retinoic acid response element. Proc. Natl. Acad. Sci. USA 1991, 88, 10138–10142. [Google Scholar] [CrossRef] [PubMed]
  124. Leroy, P.; Krust, A.; Zelent, A.; Mendelsohn, C.; Garnier, J.M.; Kastner, P.; Dierich, A.; Chambon, P. Multiple isoforms of the mouse retinoic acid receptor alpha are generated by alternative splicing and differential induction by retinoic acid. EMBO J. 1991, 10, 59–69. [Google Scholar] [CrossRef] [PubMed]
  125. Lehmann, J.M.; Zhang, X.K.; Pfahl, M. RAR gamma 2 expression is regulated through a retinoic acid response element embedded in Sp1 sites. Mol. Cell Biol. 1992, 12, 2976–2985. [Google Scholar] [CrossRef]
  126. Citron, B.A.; Dennis, J.S.; Zeitlin, R.S.; Echeverria, V. Transcription factor Sp1 dysregulation in Alzheimer’s disease. J. Neurosci. Res. 2008, 86, 2499–2504. [Google Scholar] [CrossRef]
  127. Xu, L.; Glass, C.K.; Rosenfeld, M.G. Coactivator and corepressor complexes in nuclear receptor function. Curr. Opin. Genet. Dev. 1999, 9, 140–147. [Google Scholar] [CrossRef]
  128. Gurevich, I.; Aneskievich, B.J. Liganded RARalpha and RARgamma interact with but are repressed by TNIP1. Biochem. Biophys. Res. Commun. 2009, 389, 409–414. [Google Scholar] [CrossRef]
  129. Chen, J.D.; Evans, R.M. A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature 1995, 377, 454–457. [Google Scholar] [CrossRef]
  130. Grignani, F.; De Matteis, S.; Nervi, C.; Tomassoni, L.; Gelmetti, V.; Cioce, M.; Fanelli, M.; Ruthardt, M.; Ferrara, F.F.; Zamir, I.; et al. Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia. Nature 1998, 391, 815–818. [Google Scholar] [CrossRef]
  131. Martin, P.J.; Delmotte, M.H.; Formstecher, P.; Lefebvre, P. PLZF is a negative regulator of retinoic acid receptor transcriptional activity. Nucl. Recept. 2003, 1, 6. [Google Scholar] [CrossRef]
  132. Flores, A.M.; Gurevich, I.; Zhang, C.; Ramirez, V.P.; Devens, T.R.; Aneskievich, B.J. TNIP1 is a corepressor of agonist-bound PPARs. Arch. Biochem. Biophys. 2011, 516, 58–66. [Google Scholar] [CrossRef]
  133. Li, H.J.; Haque, Z.K.; Chen, A.; Mendelsohn, M. RIF-1, a novel nuclear receptor corepressor that associates with the nuclear matrix. J. Cell Biochem. 2007, 102, 1021–1035. [Google Scholar] [CrossRef]
  134. Fernandes, I.; Bastien, Y.; Wai, T.; Nygard, K.; Lin, R.; Cormier, O.; Lee, H.S.; Eng, F.; Bertos, N.R.; Pelletier, N.; et al. Ligand-dependent nuclear receptor corepressor LCoR functions by histone deacetylase-dependent and -independent mechanisms. Mol. Cell 2003, 11, 139–150. [Google Scholar] [CrossRef]
  135. Li, M.; Tian, X.; An, R.; Yang, M.; Zhang, Q.; Xiang, F.; Liu, H.; Wang, Y.; Xu, L.; Dong, Z. All-Trans Retinoic Acid Ameliorates the Early Experimental Cerebral Ischemia-Reperfusion Injury in Rats by Inhibiting the Loss of the Blood-Brain Barrier via the JNK/P38MAPK Signaling Pathway. Neurochem. Res. 2018, 43, 1283–1296. [Google Scholar] [CrossRef]
  136. John, A.; Kubosumi, A.; Reddy, P.H. Mitochondrial MicroRNAs in Aging and Neurodegenerative Diseases. Cells 2020, 9, 1345. [Google Scholar] [CrossRef]
  137. Reed, T.T.; Pierce, W.M.; Markesbery, W.R.; Butterfield, D.A. Proteomic identification of HNE-bound proteins in early Alzheimer disease: Insights into the role of lipid peroxidation in the progression of AD. Brain Res. 2009, 1274, 66–76. [Google Scholar] [CrossRef]
  138. Bastien, J.; Rochette-Egly, C. Nuclear retinoid receptors and the transcription of retinoid-target genes. Gene 2004, 328, 1–16. [Google Scholar] [CrossRef]
  139. Petkovich, M.; Chambon, P. Retinoic acid receptors at 35 years. J. Mol. Endocrinol. 2022, 69, T13–T24. [Google Scholar] [CrossRef]
  140. Siddikuzzaman; Grace, V.M. Antioxidant potential of all-trans retinoic acid (ATRA) and enhanced activity of liposome encapsulated ATRA against inflammation and tumor-directed angiogenesis. Immunopharmacol. Immunotoxicol. 2013, 35, 164–173. [Google Scholar] [CrossRef]
  141. Tesoriere, L.; Ciaccio, M.; Bongiorno, A.; Riccio, A.; Pintaudi, A.M.; Livrea, M.A. Antioxidant activity of all-trans-retinol in homogeneous solution and in phosphatidylcholine liposomes. Arch. Biochem. Biophys. 1993, 307, 217–223. [Google Scholar] [CrossRef] [PubMed]
  142. Bohm, F.; Edge, R.; Truscott, G. Interactions of dietary carotenoids with activated (singlet) oxygen and free radicals: Potential effects for human health. Mol. Nutr. Food Res. 2012, 56, 205–216. [Google Scholar] [CrossRef] [PubMed]
  143. Glorieux, C.; Zamocky, M.; Sandoval, J.M.; Verrax, J.; Calderon, P.B. Regulation of catalase expression in healthy and cancerous cells. Free Radic. Biol. Med. 2015, 87, 84–97. [Google Scholar] [CrossRef]
  144. Calsolaro, V.; Edison, P. Neuroinflammation in Alzheimer’s disease: Current evidence and future directions. Alzheimers Dement. 2016, 12, 719–732. [Google Scholar] [CrossRef] [PubMed]
  145. Marttinen, M.; Takalo, M.; Natunen, T.; Wittrahm, R.; Gabbouj, S.; Kemppainen, S.; Leinonen, V.; Tanila, H.; Haapasalo, A.; Hiltunen, M. Molecular Mechanisms of Synaptotoxicity and Neuroinflammation in Alzheimer’s Disease. Front. Neurosci. 2018, 12, 963. [Google Scholar] [CrossRef] [PubMed]
  146. Swardfager, W.; Lanctot, K.; Rothenburg, L.; Wong, A.; Cappell, J.; Herrmann, N. A meta-analysis of cytokines in Alzheimer’s disease. Biol. Psychiatry 2010, 68, 930–941. [Google Scholar] [CrossRef]
  147. Liu, G.H.; Qu, J.; Shen, X. NF-kappaB/p65 antagonizes Nrf2-ARE pathway by depriving CBP from Nrf2 and facilitating recruitment of HDAC3 to MafK. Biochim. Biophys. Acta 2008, 1783, 713–727. [Google Scholar] [CrossRef]
  148. Cuadrado, A.; Manda, G.; Hassan, A.; Alcaraz, M.J.; Barbas, C.; Daiber, A.; Ghezzi, P.; Leon, R.; Lopez, M.G.; Oliva, B.; et al. Transcription Factor NRF2 as a Therapeutic Target for Chronic Diseases: A Systems Medicine Approach. Pharmacol. Rev. 2018, 70, 348–383. [Google Scholar] [CrossRef]
  149. Sierra-Mondragon, E.; Molina-Jijon, E.; Namorado-Tonix, C.; Rodriguez-Munoz, R.; Pedraza-Chaverri, J.; Reyes, J.L. All-trans retinoic acid ameliorates inflammatory response mediated by TLR4/NF-kappaB during initiation of diabetic nephropathy. J. Nutr. Biochem. 2018, 60, 47–60. [Google Scholar] [CrossRef]
  150. Rafa, H.; Benkhelifa, S.; AitYounes, S.; Saoula, H.; Belhadef, S.; Belkhelfa, M.; Boukercha, A.; Toumi, R.; Soufli, I.; Morales, O.; et al. All-Trans Retinoic Acid Modulates TLR4/NF-kappaB Signaling Pathway Targeting TNF-alpha and Nitric Oxide Synthase 2 Expression in Colonic Mucosa during Ulcerative Colitis and Colitis Associated Cancer. Mediat. Inflamm. 2017, 2017, 7353252. [Google Scholar] [CrossRef]
  151. Wang, R.; Chen, S.; Liu, Y.; Diao, S.; Xue, Y.; You, X.; Park, E.A.; Liao, F.F. All-trans-retinoic acid reduces BACE1 expression under inflammatory conditions via modulation of nuclear factor kappaB (NFkappaB) signaling. J. Biol. Chem. 2015, 290, 22532–22542. [Google Scholar] [CrossRef]
  152. Kirchmeyer, M.; Koufany, M.; Sebillaud, S.; Netter, P.; Jouzeau, J.Y.; Bianchi, A. All-trans retinoic acid suppresses interleukin-6 expression in interleukin-1-stimulated synovial fibroblasts by inhibition of ERK1/2 pathway independently of RAR activation. Arthritis Res. Ther. 2008, 10, R141. [Google Scholar] [CrossRef] [PubMed]
  153. Mehta, K.; McQueen, T.; Tucker, S.; Pandita, R.; Aggarwal, B.B. Inhibition by all-trans-retinoic acid of tumor necrosis factor and nitric oxide production by peritoneal macrophages. J. Leukoc. Biol. 1994, 55, 336–342. [Google Scholar] [CrossRef] [PubMed]
  154. Nozaki, Y.; Yamagata, T.; Sugiyama, M.; Ikoma, S.; Kinoshita, K.; Funauchi, M. Anti-inflammatory effect of all-trans-retinoic acid in inflammatory arthritis. Clin. Immunol. 2006, 119, 272–279. [Google Scholar] [CrossRef] [PubMed]
  155. Zitnik, R.J.; Kotloff, R.M.; Latifpour, J.; Zheng, T.; Whiting, N.L.; Schwalb, J.; Elias, J.A. Retinoic acid inhibition of IL-1-induced IL-6 production by human lung fibroblasts. J. Immunol. 1994, 152, 1419–1427. [Google Scholar] [CrossRef]
  156. Austenaa, L.M.; Carlsen, H.; Ertesvag, A.; Alexander, G.; Blomhoff, H.K.; Blomhoff, R. Vitamin A status significantly alters nuclear factor-kappaB activity assessed by in vivo imaging. FASEB J. 2004, 18, 1255–1257. [Google Scholar] [CrossRef]
  157. Hong, K.; Zhang, Y.; Guo, Y.; Xie, J.; Wang, J.; He, X.; Lu, N.; Bai, A. All-trans retinoic acid attenuates experimental colitis through inhibition of NF-kappaB signaling. Immunol. Lett. 2014, 162, 34–40. [Google Scholar] [CrossRef]
  158. Osama, A.; Zhang, J.; Yao, J.; Yao, X.; Fang, J. Nrf2: A dark horse in Alzheimer’s disease treatment. Ageing Res. Rev. 2020, 64, 101206. [Google Scholar] [CrossRef]
  159. Wang, X.J.; Hayes, J.D.; Henderson, C.J.; Wolf, C.R. Identification of retinoic acid as an inhibitor of transcription factor Nrf2 through activation of retinoic acid receptor alpha. Proc. Natl. Acad. Sci. USA 2007, 104, 19589–19594. [Google Scholar] [CrossRef]
  160. Wu, J.; Wang, H.; Tang, X. Rexinoid inhibits Nrf2-mediated transcription through retinoid X receptor alpha. Biochem. Biophys. Res. Commun. 2014, 452, 554–559. [Google Scholar] [CrossRef]
  161. Kobayashi, A.; Kang, M.I.; Watai, Y.; Tong, K.I.; Shibata, T.; Uchida, K.; Yamamoto, M. Oxidative and electrophilic stresses activate Nrf2 through inhibition of ubiquitination activity of Keap1. Mol. Cell Biol. 2006, 26, 221–229. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, H.; Liu, K.; Geng, M.; Gao, P.; Wu, X.; Hai, Y.; Li, Y.; Li, Y.; Luo, L.; Hayes, J.D.; et al. RXRalpha inhibits the NRF2-ARE signaling pathway through a direct interaction with the Neh7 domain of NRF2. Cancer Res. 2013, 73, 3097–3108. [Google Scholar] [CrossRef] [PubMed]
  163. Valenzuela, M.; Glorieux, C.; Stockis, J.; Sid, B.; Sandoval, J.M.; Felipe, K.B.; Kviecinski, M.R.; Verrax, J.; Buc Calderon, P. Retinoic acid synergizes ATO-mediated cytotoxicity by precluding Nrf2 activity in AML cells. Br. J. Cancer 2014, 111, 874–882. [Google Scholar] [CrossRef] [PubMed]
  164. Shearer, K.D.; Stoney, P.N.; Morgan, P.J.; McCaffery, P.J. A vitamin for the brain. Trends Neurosci. 2012, 35, 733–741. [Google Scholar] [CrossRef]
  165. Sandberg, M.; Patil, J.; D’Angelo, B.; Weber, S.G.; Mallard, C. NRF2-regulation in brain health and disease: Implication of cerebral inflammation. Neuropharmacology 2014, 79, 298–306. [Google Scholar] [CrossRef]
  166. Uruno, A.; Matsumaru, D.; Ryoke, R.; Saito, R.; Kadoguchi, S.; Saigusa, D.; Saito, T.; Saido, T.C.; Kawashima, R.; Yamamoto, M. Nrf2 Suppresses Oxidative Stress and Inflammation in App Knock-In Alzheimer’s Disease Model Mice. Mol. Cell Biol. 2020, 40, e00467-19. [Google Scholar] [CrossRef]
  167. Lipton, S.A.; Rezaie, T.; Nutter, A.; Lopez, K.M.; Parker, J.; Kosaka, K.; Satoh, T.; McKercher, S.R.; Masliah, E.; Nakanishi, N. Therapeutic advantage of pro-electrophilic drugs to activate the Nrf2/ARE pathway in Alzheimer’s disease models. Cell Death Dis. 2016, 7, e2499. [Google Scholar] [CrossRef]
  168. Rojo, A.I.; Pajares, M.; Rada, P.; Nunez, A.; Nevado-Holgado, A.J.; Killik, R.; Van Leuven, F.; Ribe, E.; Lovestone, S.; Yamamoto, M.; et al. NRF2 deficiency replicates transcriptomic changes in Alzheimer’s patients and worsens APP and TAU pathology. Redox Biol. 2017, 13, 444–451. [Google Scholar] [CrossRef]
  169. Eisenberg-Bord, M.; Schuldiner, M. Ground control to major TOM: Mitochondria-nucleus communication. FEBS J. 2017, 284, 196–210. [Google Scholar] [CrossRef]
  170. Terada, K.; Kanazawa, M.; Yano, M.; Hanson, B.; Hoogenraad, N.; Mori, M. Participation of the import receptor Tom20 in protein import into mammalian mitochondria: Analyses in vitro and in cultured cells. FEBS Lett. 1997, 403, 309–312. [Google Scholar] [CrossRef]
  171. Rapaport, D.; Neupert, W.; Lill, R. Mitochondrial protein import. Tom40 plays a major role in targeting and translocation of preproteins by forming a specific binding site for the presequence. J. Biol. Chem. 1997, 272, 18725–18731. [Google Scholar] [CrossRef] [PubMed]
  172. Chai, Y.L.; Xing, H.; Chong, J.R.; Francis, P.T.; Ballard, C.G.; Chen, C.P.; Lai, M.K.P. Mitochondrial Translocase of the Outer Membrane Alterations May Underlie Dysfunctional Oxidative Phosphorylation in Alzheimer’s Disease. J. Alzheimers Dis. 2018, 61, 793–801. [Google Scholar] [CrossRef]
  173. Wright, G.; Terada, K.; Yano, M.; Sergeev, I.; Mori, M. Oxidative stress inhibits the mitochondrial import of preproteins and leads to their degradation. Exp. Cell Res. 2001, 263, 107–117. [Google Scholar] [CrossRef]
  174. Hansson Petersen, C.A.; Alikhani, N.; Behbahani, H.; Wiehager, B.; Pavlov, P.F.; Alafuzoff, I.; Leinonen, V.; Ito, A.; Winblad, B.; Glaser, E.; et al. The amyloid beta-peptide is imported into mitochondria via the TOM import machinery and localized to mitochondrial cristae. Proc. Natl. Acad. Sci. USA 2008, 105, 13145–13150. [Google Scholar] [CrossRef] [PubMed]
  175. Du, H.; Guo, L.; Yan, S.; Sosunov, A.A.; McKhann, G.M.; Yan, S.S. Early deficits in synaptic mitochondria in an Alzheimer’s disease mouse model. Proc. Natl. Acad. Sci. USA 2010, 107, 18670–18675. [Google Scholar] [CrossRef] [PubMed]
  176. Du, H.; Guo, L.; Yan, S.S. Synaptic mitochondrial pathology in Alzheimer’s disease. Antioxid. Redox Signal 2012, 16, 1467–1475. [Google Scholar] [CrossRef]
  177. Gao, S.; Hu, J. Mitochondrial Fusion: The Machineries In and Out. Trends Cell Biol. 2021, 31, 62–74. [Google Scholar] [CrossRef]
  178. Wang, X.; Su, B.; Lee, H.G.; Li, X.; Perry, G.; Smith, M.A.; Zhu, X. Impaired balance of mitochondrial fission and fusion in Alzheimer’s disease. J. Neurosci. 2009, 28, 9090–9103. [Google Scholar] [CrossRef]
  179. Perez, M.J.; Ponce, D.P.; Osorio-Fuentealba, C.; Behrens, M.I.; Quintanilla, R.A. Mitochondrial Bioenergetics Is Altered in Fibroblasts from Patients with Sporadic Alzheimer’s Disease. Front. Neurosci. 2017, 11, 553. [Google Scholar] [CrossRef]
  180. Reddy, P.H.; Reddy, T.P.; Manczak, M.; Calkins, M.J.; Shirendeb, U.; Mao, P. Dynamin-related protein 1 and mitochondrial fragmentation in neurodegenerative diseases. Brain Res. Rev. 2011, 67, 103–118. [Google Scholar] [CrossRef]
  181. Lv, F.; Yang, X.; Cui, C.; Su, C. Exogenous expression of Drp1 plays neuroprotective roles in the Alzheimer’s disease in the Abeta42 transgenic Drosophila model. PLoS ONE 2017, 12, e0176183. [Google Scholar] [CrossRef] [PubMed]
  182. Itoh, K.; Murata, D.; Kato, T.; Yamada, T.; Araki, Y.; Saito, A.; Adachi, Y.; Igarashi, A.; Li, S.; Pletnikov, M.; et al. Brain-specific Drp1 regulates postsynaptic endocytosis and dendrite formation independently of mitochondrial division. Elife 2019, 8, e44739. [Google Scholar] [CrossRef] [PubMed]
  183. Chidipi, B.; Shah, S.I.; Reiser, M.; Kanithi, M.; Garces, A.; Cha, B.J.; Ullah, G.; Noujaim, S.F. All-Trans Retinoic Acid Increases DRP1 Levels and Promotes Mitochondrial Fission. Cells 2021, 10, 1202. [Google Scholar] [CrossRef] [PubMed]
  184. Acquaah-Mensah, G.K.; Agu, N.; Khan, T.; Gardner, A. A regulatory role for the insulin- and BDNF-linked RORA in the hippocampus: Implications for Alzheimer’s disease. J. Alzheimers Dis. 2015, 44, 827–838. [Google Scholar] [CrossRef]
  185. Stehlin-Gaon, C.; Willmann, D.; Zeyer, D.; Sanglier, S.; Van Dorsselaer, A.; Renaud, J.P.; Moras, D.; Schule, R. All-trans retinoic acid is a ligand for the orphan nuclear receptor ROR beta. Nat. Struct. Biol. 2003, 10, 820–825. [Google Scholar] [CrossRef]
  186. Baek, S.H.; Park, S.J.; Jeong, J.I.; Kim, S.H.; Han, J.; Kyung, J.W.; Baik, S.H.; Choi, Y.; Choi, B.Y.; Park, J.S.; et al. Inhibition of Drp1 Ameliorates Synaptic Depression, Abeta Deposition, and Cognitive Impairment in an Alzheimer’s Disease Model. J. Neurosci. 2017, 37, 5099–5110. [Google Scholar] [CrossRef]
  187. Joshi, A.U.; Saw, N.L.; Shamloo, M.; Mochly-Rosen, D. Drp1/Fis1 interaction mediates mitochondrial dysfunction, bioenergetic failure and cognitive decline in Alzheimer’s disease. Oncotarget 2018, 9, 6128–6143. [Google Scholar] [CrossRef] [PubMed]
  188. Grohm, J.; Kim, S.W.; Mamrak, U.; Tobaben, S.; Cassidy-Stone, A.; Nunnari, J.; Plesnila, N.; Culmsee, C. Inhibition of Drp1 provides neuroprotection in vitro and in vivo. Cell Death Differ. 2012, 19, 1446–1458. [Google Scholar] [CrossRef]
  189. Rosdah, A.A.; Smiles, W.J.; Oakhill, J.S.; Scott, J.W.; Langendorf, C.G.; Delbridge, L.M.D.; Holien, J.K.; Lim, S.Y. New perspectives on the role of Drp1 isoforms in regulating mitochondrial pathophysiology. Pharmacol. Ther. 2020, 213, 107594. [Google Scholar] [CrossRef]
  190. Abtahi, S.L.; Masoudi, R.; Haddadi, M. The distinctive role of tau and amyloid beta in mitochondrial dysfunction through alteration in Mfn2 and Drp1 mRNA Levels: A comparative study in Drosophila melanogaster. Gene 2020, 754, 144854. [Google Scholar] [CrossRef] [PubMed]
  191. Manczak, M.; Kandimalla, R.; Fry, D.; Sesaki, H.; Reddy, P.H. Protective effects of reduced dynamin-related protein 1 against amyloid beta-induced mitochondrial dysfunction and synaptic damage in Alzheimer’s disease. Hum. Mol. Genet. 2016, 25, 5148–5166. [Google Scholar] [CrossRef]
  192. Manczak, M.; Reddy, P.H. Abnormal interaction between the mitochondrial fission protein Drp1 and hyperphosphorylated tau in Alzheimer’s disease neurons: Implications for mitochondrial dysfunction and neuronal damage. Hum. Mol. Genet. 2012, 21, 2538–2547. [Google Scholar] [CrossRef] [PubMed]
  193. Manczak, M.; Calkins, M.J.; Reddy, P.H. Impaired mitochondrial dynamics and abnormal interaction of amyloid beta with mitochondrial protein Drp1 in neurons from patients with Alzheimer’s disease: Implications for neuronal damage. Hum. Mol. Genet. 2011, 20, 2495–2509. [Google Scholar] [CrossRef] [PubMed]
  194. Yan, J.; Liu, X.H.; Han, M.Z.; Wang, Y.M.; Sun, X.L.; Yu, N.; Li, T.; Su, B.; Chen, Z.Y. Blockage of GSK3beta-mediated Drp1 phosphorylation provides neuroprotection in neuronal and mouse models of Alzheimer’s disease. Neurobiol. Aging 2015, 36, 211–227. [Google Scholar] [CrossRef] [PubMed]
  195. Kim, D.I.; Lee, K.H.; Gabr, A.A.; Choi, G.E.; Kim, J.S.; Ko, S.H.; Han, H.J. Abeta-Induced Drp1 phosphorylation through Akt activation promotes excessive mitochondrial fission leading to neuronal apoptosis. Biochim. Biophys. Acta 2016, 1863, 2820–2834. [Google Scholar] [CrossRef]
  196. DuBoff, B.; Gotz, J.; Feany, M.B. Tau promotes neurodegeneration via DRP1 mislocalization in vivo. Neuron 2012, 75, 618–632. [Google Scholar] [CrossRef]
  197. Kuruva, C.S.; Manczak, M.; Yin, X.; Ogunmokun, G.; Reddy, A.P.; Reddy, P.H. Aqua-soluble DDQ reduces the levels of Drp1 and Abeta and inhibits abnormal interactions between Abeta and Drp1 and protects Alzheimer’s disease neurons from Abeta- and Drp1-induced mitochondrial and synaptic toxicities. Hum. Mol. Genet. 2017, 26, 3375–3395. [Google Scholar] [CrossRef]
  198. Kandimalla, R.; Manczak, M.; Fry, D.; Suneetha, Y.; Sesaki, H.; Reddy, P.H. Reduced dynamin-related protein 1 protects against phosphorylated Tau-induced mitochondrial dysfunction and synaptic damage in Alzheimer’s disease. Hum. Mol. Genet. 2016, 25, 4881–4897. [Google Scholar] [CrossRef]
  199. Wei, Y.; Qian, M. Case Report: A Novel de novo Mutation in DNM1L Presenting With Developmental Delay, Ataxia, and Peripheral Neuropathy. Front. Pediatr. 2021, 9, 604105. [Google Scholar] [CrossRef] [PubMed]
  200. Kandimalla, R.; Reddy, P.H. Multiple faces of dynamin-related protein 1 and its role in Alzheimer’s disease pathogenesis. Biochim. Biophys. Acta 2016, 1862, 814–828. [Google Scholar] [CrossRef]
  201. Seto, E.; Yoshida, M. Erasers of histone acetylation: The histone deacetylase enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef] [PubMed]
  202. Shukla, S.; Tekwani, B.L. Histone Deacetylases Inhibitors in Neurodegenerative Diseases, Neuroprotection and Neuronal Differentiation. Front. Pharmacol. 2020, 11, 537. [Google Scholar] [CrossRef]
  203. Xu, K.; Dai, X.L.; Huang, H.C.; Jiang, Z.F. Targeting HDACs: A promising therapy for Alzheimer’s disease. Oxid. Med. Cell. Longev. 2011, 2011, 143269. [Google Scholar] [CrossRef]
  204. Kilgore, M.; Miller, C.A.; Fass, D.M.; Hennig, K.M.; Haggarty, S.J.; Sweatt, J.D.; Rumbaugh, G. Inhibitors of class 1 histone deacetylases reverse contextual memory deficits in a mouse model of Alzheimer’s disease. Neuropsychopharmacology 2010, 35, 870–880. [Google Scholar] [CrossRef]
  205. Ray, P.D.; Huang, B.W.; Tsuji, Y. Coordinated regulation of Nrf2 and histone H3 serine 10 phosphorylation in arsenite-activated transcription of the human heme oxygenase-1 gene. Biochim. Biophys. Acta 2015, 1849, 1277–1288. [Google Scholar] [CrossRef]
  206. Correa, F.; Mallard, C.; Nilsson, M.; Sandberg, M. Activated microglia decrease histone acetylation and Nrf2-inducible anti-oxidant defence in astrocytes: Restoring effects of inhibitors of HDACs, p38 MAPK and GSK3beta. Neurobiol. Dis. 2011, 44, 142–151. [Google Scholar] [CrossRef]
  207. Yu, S.; Khor, T.O.; Cheung, K.L.; Li, W.; Wu, T.Y.; Huang, Y.; Foster, B.A.; Kan, Y.W.; Kong, A.N. Nrf2 expression is regulated by epigenetic mechanisms in prostate cancer of TRAMP mice. PLoS ONE 2010, 5, e8579. [Google Scholar] [CrossRef]
  208. Guo, Y.; Yu, S.; Zhang, C.; Kong, A.N. Epigenetic regulation of Keap1-Nrf2 signaling. Free Radic. Biol. Med. 2015, 88, 337–349. [Google Scholar] [CrossRef]
  209. Silva-Palacios, A.; Ostolga-Chavarria, M.; Zazueta, C.; Konigsberg, M. Nrf2: Molecular and epigenetic regulation during aging. Ageing Res. Rev. 2018, 47, 31–40. [Google Scholar] [CrossRef]
  210. Graff, J.; Rei, D.; Guan, J.S.; Wang, W.Y.; Seo, J.; Hennig, K.M.; Nieland, T.J.; Fass, D.M.; Kao, P.F.; Kahn, M.; et al. An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature 2012, 483, 222–226. [Google Scholar] [CrossRef] [PubMed]
  211. Coppieters, N.; Dieriks, B.V.; Lill, C.; Faull, R.L.; Curtis, M.A.; Dragunow, M. Global changes in DNA methylation and hydroxymethylation in Alzheimer’s disease human brain. Neurobiol. Aging 2014, 35, 1334–1344. [Google Scholar] [CrossRef]
  212. Karagiannis, T.C.; Ververis, K. Potential of chromatin modifying compounds for the treatment of Alzheimer’s disease. Pathobiol. Aging Age Relat. Dis. 2012, 2, 14980. [Google Scholar] [CrossRef]
  213. Coppede, F. The potential of epigenetic therapies in neurodegenerative diseases. Front. Genet. 2014, 5, 220. [Google Scholar] [CrossRef]
  214. Schueller, E.; Paiva, I.; Blanc, F.; Wang, X.L.; Cassel, J.C.; Boutillier, A.L.; Bousiges, O. Dysregulation of histone acetylation pathways in hippocampus and frontal cortex of Alzheimer’s disease patients. Eur. Neuropsychopharmacol. 2020, 33, 101–116. [Google Scholar] [CrossRef]
  215. Pellegrini, C.; Pirazzini, C.; Sala, C.; Sambati, L.; Yusipov, I.; Kalyakulina, A.; Ravaioli, F.; Kwiatkowska, K.M.; Durso, D.F.; Ivanchenko, M.; et al. A Meta-Analysis of Brain DNA Methylation Across Sex, Age, and Alzheimer’s Disease Points for Accelerated Epigenetic Aging in Neurodegeneration. Front. Aging Neurosci. 2021, 13, 639428. [Google Scholar] [CrossRef]
  216. Hou, N.; Ren, L.; Gong, M.; Bi, Y.; Gu, Y.; Dong, Z.; Liu, Y.; Chen, J.; Li, T. Vitamin A deficiency impairs spatial learning and memory: The mechanism of abnormal CBP-dependent histone acetylation regulated by retinoic acid receptor alpha. Mol. Neurobiol. 2015, 51, 633–647. [Google Scholar] [CrossRef]
  217. Fischer, A.; Sananbenesi, F.; Wang, X.; Dobbin, M.; Tsai, L.H. Recovery of learning and memory is associated with chromatin remodelling. Nature 2007, 447, 178–182. [Google Scholar] [CrossRef]
  218. Ricobaraza, A.; Cuadrado-Tejedor, M.; Marco, S.; Perez-Otano, I.; Garcia-Osta, A. Phenylbutyrate rescues dendritic spine loss associated with memory deficits in a mouse model of Alzheimer disease. Hippocampus 2012, 22, 1040–1050. [Google Scholar] [CrossRef]
  219. Govindarajan, N.; Agis-Balboa, R.C.; Walter, J.; Sananbenesi, F.; Fischer, A. Sodium butyrate improves memory function in an Alzheimer’s disease mouse model when administered at an advanced stage of disease progression. J. Alzheimers Dis. 2011, 26, 187–197. [Google Scholar] [CrossRef]
  220. Francis, Y.I.; Fa, M.; Ashraf, H.; Zhang, H.; Staniszewski, A.; Latchman, D.S.; Arancio, O. Dysregulation of histone acetylation in the APP/PS1 mouse model of Alzheimer’s disease. J. Alzheimers Dis. 2009, 18, 131–139. [Google Scholar] [CrossRef]
  221. Zhang, Z.Y.; Schluesener, H.J. Oral administration of histone deacetylase inhibitor MS-275 ameliorates neuroinflammation and cerebral amyloidosis and improves behavior in a mouse model. J. Neuropathol. Exp. Neurol. 2013, 72, 178–185. [Google Scholar] [CrossRef]
  222. Sung, Y.M.; Lee, T.; Yoon, H.; DiBattista, A.M.; Song, J.M.; Sohn, Y.; Moffat, E.I.; Turner, R.S.; Jung, M.; Kim, J.; et al. Mercaptoacetamide-based class II HDAC inhibitor lowers Abeta levels and improves learning and memory in a mouse model of Alzheimer’s disease. Exp. Neurol. 2013, 239, 192–201. [Google Scholar] [CrossRef]
  223. De Simone, A.; Milelli, A. Histone Deacetylase Inhibitors as Multitarget Ligands: New Players in Alzheimer’s Disease Drug Discovery? ChemMedChem 2019, 14, 1067–1073. [Google Scholar] [CrossRef] [PubMed]
  224. Burns, A.M.; Graff, J. Cognitive epigenetic priming: Leveraging histone acetylation for memory amelioration. Curr. Opin. Neurobiol. 2021, 67, 75–84. [Google Scholar] [CrossRef]
  225. Han, Y.; Chen, L.; Guo, Y.; Wang, C.; Zhang, C.; Kong, L.; Ma, H. Class I HDAC Inhibitor Improves Synaptic Proteins and Repairs Cytoskeleton Through Regulating Synapse-Related Genes In vitro and In vivo. Front. Aging Neurosci. 2020, 12, 619866. [Google Scholar] [CrossRef]
  226. Kitahara, M.; Inoue, T.; Mani, H.; Takamatsu, Y.; Ikegami, R.; Tohyama, H.; Maejima, H. Exercise and pharmacological inhibition of histone deacetylase improves cognitive function accompanied by an increase of gene expressions crucial for neuronal plasticity in the hippocampus. Neurosci. Lett. 2021, 749, 135749. [Google Scholar] [CrossRef] [PubMed]
  227. Yang, S.S.; Zhang, R.; Wang, G.; Zhang, Y.F. The development prospection of HDAC inhibitors as a potential therapeutic direction in Alzheimer’s disease. Transl. Neurodegener. 2017, 6, 19. [Google Scholar] [CrossRef]
  228. Sodhi, R.K.; Singh, N. Retinoids as potential targets for Alzheimer’s disease. Pharmacol. Biochem. Behav. 2014, 120, 117–123. [Google Scholar] [CrossRef] [PubMed]
  229. Timmermann, S.; Lehrmann, H.; Polesskaya, A.; Harel-Bellan, A. Histone acetylation and disease. Cell Mol. Life Sci. 2001, 58, 728–736. [Google Scholar] [CrossRef]
  230. Moon, M.; Um, S.J.; Kim, E.J. CAC1 negatively regulates RARalpha activity through cooperation with HDAC. Biochem. Biophys. Res. Commun. 2012, 427, 41–46. [Google Scholar] [CrossRef]
  231. Thiagarajan, D.; Ananthakrishnan, R.; Zhang, J.; O’Shea, K.M.; Quadri, N.; Li, Q.; Sas, K.; Jing, X.; Rosario, R.; Pennathur, S.; et al. Aldose Reductase Acts as a Selective Derepressor of PPARgamma and the Retinoic Acid Receptor. Cell Rep. 2016, 15, 181–196. [Google Scholar] [CrossRef] [PubMed]
  232. Kleinschmidt, M.A.; Streubel, G.; Samans, B.; Krause, M.; Bauer, U.M. The protein arginine methyltransferases CARM1 and PRMT1 cooperate in gene regulation. Nucleic Acids Res. 2008, 36, 3202–3213. [Google Scholar] [CrossRef] [PubMed]
  233. Greer, E.L.; Shi, Y. Histone methylation: A dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 2012, 13, 343–357. [Google Scholar] [CrossRef]
  234. Legrand, N.; Bretscher, C.L.; Zielke, S.; Wilke, B.; Daude, M.; Fritz, B.; Diederich, W.E.; Adhikary, T. PPARbeta/delta recruits NCOR and regulates transcription reinitiation of ANGPTL4. Nucleic Acids Res. 2019, 47, 9573–9591. [Google Scholar] [CrossRef]
  235. Spencer, T.E.; Jenster, G.; Burcin, M.M.; Allis, C.D.; Zhou, J.; Mizzen, C.A.; McKenna, N.J.; Onate, S.A.; Tsai, S.Y.; Tsai, M.J.; et al. Steroid receptor coactivator-1 is a histone acetyltransferase. Nature 1997, 389, 194–198. [Google Scholar] [CrossRef] [PubMed]
  236. Tan, N.S.; Vazquez-Carrera, M.; Montagner, A.; Sng, M.K.; Guillou, H.; Wahli, W. Transcriptional control of physiological and pathological processes by the nuclear receptor PPARbeta/delta. Prog. Lipid Res. 2016, 64, 98–122. [Google Scholar] [CrossRef]
  237. Sun, Z.; Chin, Y.E.; Zhang, D.D. Acetylation of Nrf2 by p300/CBP augments promoter-specific DNA binding of Nrf2 during the antioxidant response. Mol. Cell Biol. 2009, 29, 2658–2672. [Google Scholar] [CrossRef]
  238. Bahn, G.; Jo, D.G. Therapeutic Approaches to Alzheimer’s Disease Through Modulation of NRF2. Neuromolecular Med. 2019, 21, 1–11. [Google Scholar] [CrossRef]
  239. Pu, J.; Chen, D.; Tian, G.; He, J.; Huang, Z.; Zheng, P.; Mao, X.; Yu, J.; Luo, J.; Luo, Y.; et al. All-Trans Retinoic Acid Attenuates Transmissible Gastroenteritis Virus-Induced Apoptosis in IPEC-J2 Cells via Inhibiting ROS-Mediated P(38)MAPK Signaling Pathway. Antioxidants 2022, 11, 345. [Google Scholar] [CrossRef]
  240. Evans, R.M.; Mangelsdorf, D.J. Nuclear Receptors, RXR, and the Big Bang. Cell 2014, 157, 255–266. [Google Scholar] [CrossRef]
  241. Rochette-Egly, C. Retinoic acid signaling and mouse embryonic stem cell differentiation: Cross talk between genomic and non-genomic effects of RA. Biochim. Biophys. Acta 2015, 1851, 66–75. [Google Scholar] [CrossRef]
  242. Chen, L.; Lau, A.G.; Sarti, F. Synaptic retinoic acid signaling and homeostatic synaptic plasticity. Neuropharmacology 2014, 78, 3–12. [Google Scholar] [CrossRef]
  243. Arendt, K.L.; Zhang, Z.; Ganesan, S.; Hintze, M.; Shin, M.M.; Tang, Y.; Cho, A.; Graef, I.A.; Chen, L. Calcineurin mediates homeostatic synaptic plasticity by regulating retinoic acid synthesis. Proc. Natl. Acad. Sci. USA 2015, 112, E5744–E5752. [Google Scholar] [CrossRef] [PubMed]
  244. Lin, Y.L.; Persaud, S.D.; Nhieu, J.; Wei, L.N. Cellular Retinoic Acid-Binding Protein 1 Modulates Stem Cell Proliferation to Affect Learning and Memory in Male Mice. Endocrinology 2017, 158, 3004–3014. [Google Scholar] [CrossRef]
  245. Nhieu, J.; Lin, Y.L.; Wei, L.N. CRABP1 in Non-Canonical Activities of Retinoic Acid in Health and Diseases. Nutrients 2022, 14, 1528. [Google Scholar] [CrossRef] [PubMed]
  246. Theendakara, V.; Patent, A.; Peters Libeu, C.A.; Philpot, B.; Flores, S.; Descamps, O.; Poksay, K.S.; Zhang, Q.; Cailing, G.; Hart, M.; et al. Neuroprotective Sirtuin ratio reversed by ApoE4. Proc. Natl. Acad. Sci. USA 2013, 110, 18303–18308. [Google Scholar] [CrossRef] [PubMed]
  247. Zhao, F.; Zhang, J.; Chang, N. Epigenetic modification of Nrf2 by sulforaphane increases the antioxidative and anti-inflammatory capacity in a cellular model of Alzheimer’s disease. Eur. J. Pharmacol. 2018, 824, 1–10. [Google Scholar] [CrossRef]
  248. Hosseini, H.; Teimouri, M.; Shabani, M.; Koushki, M.; Babaei Khorzoughi, R.; Namvarjah, F.; Izadi, P.; Meshkani, R. Resveratrol alleviates non-alcoholic fatty liver disease through epigenetic modification of the Nrf2 signaling pathway. Int. J. Biochem. Cell Biol. 2020, 119, 105667. [Google Scholar] [CrossRef]
  249. Liu, X.; Li, H.; Wu, M.L.; Wu, J.; Sun, Y.; Zhang, K.L.; Liu, J. Resveratrol Reverses Retinoic Acid Resistance of Anaplastic Thyroid Cancer Cells via Demethylating CRABP2 Gene. Front. Endocrinol. 2019, 10, 734. [Google Scholar] [CrossRef]
  250. Chen, X.; Zhu, X.; Wei, A.; Chen, F.; Gao, Q.; Lu, K.; Jiang, Q.; Cao, W. Nrf2 epigenetic derepression induced by running exercise protects against osteoporosis. Bone Res. 2021, 9, 15. [Google Scholar] [CrossRef]
  251. Khor, T.O.; Huang, Y.; Wu, T.Y.; Shu, L.; Lee, J.; Kong, A.N. Pharmacodynamics of curcumin as DNA hypomethylation agent in restoring the expression of Nrf2 via promoter CpGs demethylation. Biochem. Pharmacol. 2011, 82, 1073–1078. [Google Scholar] [CrossRef]
  252. Donmez, G.; Outeiro, T.F. SIRT1 and SIRT2: Emerging targets in neurodegeneration. EMBO Mol. Med. 2013, 5, 344–352. [Google Scholar] [CrossRef] [PubMed]
  253. Donmez, G. The Effects of SIRT1 on Alzheimer’s Disease Models. Int. J. Alzheimers Dis. 2012, 2012, 509529. [Google Scholar] [CrossRef] [PubMed]
  254. Tang, S.; Huang, G.; Fan, W.; Chen, Y.; Ward, J.M.; Xu, X.; Xu, Q.; Kang, A.; McBurney, M.W.; Fargo, D.C.; et al. SIRT1-mediated deacetylation of CRABPII regulates cellular retinoic acid signaling and modulates embryonic stem cell differentiation. Mol. Cell 2014, 55, 843–855. [Google Scholar] [CrossRef]
  255. Lee, H.R.; Shin, H.K.; Park, S.Y.; Kim, H.Y.; Lee, W.S.; Rhim, B.Y.; Hong, K.W.; Kim, C.D. Cilostazol suppresses beta-amyloid production by activating a disintegrin and metalloproteinase 10 via the upregulation of SIRT1-coupled retinoic acid receptor-beta. J. Neurosci. Res. 2014, 92, 1581–1590. [Google Scholar] [CrossRef] [PubMed]
  256. Donmez, G.; Wang, D.; Cohen, D.E.; Guarente, L. SIRT1 suppresses beta-amyloid production by activating the alpha-secretase gene ADAM10. Cell 2010, 142, 320–332. [Google Scholar] [CrossRef]
  257. Sirchia, S.M.; Ferguson, A.T.; Sironi, E.; Subramanyan, S.; Orlandi, R.; Sukumar, S.; Sacchi, N. Evidence of epigenetic changes affecting the chromatin state of the retinoic acid receptor beta2 promoter in breast cancer cells. Oncogene 2000, 19, 1556–1563. [Google Scholar] [CrossRef]
  258. Virmani, A.K.; Rathi, A.; Zochbauer-Muller, S.; Sacchi, N.; Fukuyama, Y.; Bryant, D.; Maitra, A.; Heda, S.; Fong, K.M.; Thunnissen, F.; et al. Promoter methylation and silencing of the retinoic acid receptor-beta gene in lung carcinomas. J. Natl. Cancer Inst. 2000, 92, 1303–1307. [Google Scholar] [CrossRef]
  259. Widschwendter, M.; Berger, J.; Hermann, M.; Muller, H.M.; Amberger, A.; Zeschnigk, M.; Widschwendter, A.; Abendstein, B.; Zeimet, A.G.; Daxenbichler, G.; et al. Methylation and silencing of the retinoic acid receptor-beta2 gene in breast cancer. J. Natl. Cancer Inst. 2000, 92, 826–832. [Google Scholar] [CrossRef]
  260. Arapshian, A.; Kuppumbatti, Y.S.; Mira-y-Lopez, R. Methylation of conserved CpG sites neighboring the beta retinoic acid response element may mediate retinoic acid receptor beta gene silencing in MCF-7 breast cancer cells. Oncogene 2000, 19, 4066–4070. [Google Scholar] [CrossRef]
  261. Nakayama, T.; Watanabe, M.; Yamanaka, M.; Hirokawa, Y.; Suzuki, H.; Ito, H.; Yatani, R.; Shiraishi, T. The role of epigenetic modifications in retinoic acid receptor beta2 gene expression in human prostate cancers. Lab. Investig. 2001, 81, 1049–1057. [Google Scholar] [CrossRef]
  262. Ivanova, T.; Petrenko, A.; Gritsko, T.; Vinokourova, S.; Eshilev, E.; Kobzeva, V.; Kisseljov, F.; Kisseljova, N. Methylation and silencing of the retinoic acid receptor-beta 2 gene in cervical cancer. BMC Cancer 2002, 2, 4. [Google Scholar] [CrossRef]
  263. Zhang, Z.; Joh, K.; Yatsuki, H.; Zhao, W.; Soejima, H.; Higashimoto, K.; Noguchi, M.; Yokoyama, M.; Iwasaka, T.; Mukai, T. Retinoic acid receptor beta2 is epigenetically silenced either by DNA methylation or repressive histone modifications at the promoter in cervical cancer cells. Cancer Lett. 2007, 247, 318–327. [Google Scholar] [CrossRef] [PubMed]
  264. Huebner, H.; Strick, R.; Wachter, D.L.; Kehl, S.; Strissel, P.L.; Schneider-Stock, R.; Hartner, A.; Rascher, W.; Horn, L.C.; Beckmann, M.W.; et al. Hypermethylation and loss of retinoic acid receptor responder 1 expression in human choriocarcinoma. J. Exp. Clin. Cancer Res. 2017, 36, 165. [Google Scholar] [CrossRef]
  265. Evans, L.W.; Ferguson, B.S. Food Bioactive HDAC Inhibitors in the Epigenetic Regulation of Heart Failure. Nutrients 2018, 10, 1120. [Google Scholar] [CrossRef] [PubMed]
  266. Godoy, L.D.; Lucas, J.E.; Bender, A.J.; Romanick, S.S.; Ferguson, B.S. Targeting the epigenome: Screening bioactive compounds that regulate histone deacetylase activity. Mol. Nutr. Food Res. 2017, 61, 1600744. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Distinct roles of ATRA and Nrf2-mediated AO defenses in the hippocampus proposed during normal aging and AD. (A) The non-amyloidogenic pathway and the transcription of neuroprotective proteins is promoted by ATRA suffiency, in coordination with the RXR agonist DHA. (B) The amyloidogenic pathway is promoted by ATRA deficiency. The subsequent rise in ROS causes oxidation of the Nrf2-KEAP1 complex, resulting in a compensational increase in endogenous AO defenses.
Figure 1. Distinct roles of ATRA and Nrf2-mediated AO defenses in the hippocampus proposed during normal aging and AD. (A) The non-amyloidogenic pathway and the transcription of neuroprotective proteins is promoted by ATRA suffiency, in coordination with the RXR agonist DHA. (B) The amyloidogenic pathway is promoted by ATRA deficiency. The subsequent rise in ROS causes oxidation of the Nrf2-KEAP1 complex, resulting in a compensational increase in endogenous AO defenses.
Antioxidants 12 01921 g001
Figure 2. Theoretical detection of ATRA deficiency through gene expression analysis of human brain transcriptomics data. (A) Activation of RAR-RXR heterodimers induces increased transcription of genes that possess retinoic acid response elements (RAREs; red star). (B) In the case of ATRA sufficiency in unimpaired control brains, transcription is maintained across ATRA-sensitive genes (red stars). (C) Deficiency in ATRA leads to transcriptional repression of ATRA-sensitive genes (blue star). (D) In the case of ATRA deficiency in Alzheimer’s disease brains, transcription is decreased across ATRA-sensitive genes (blue stars).
Figure 2. Theoretical detection of ATRA deficiency through gene expression analysis of human brain transcriptomics data. (A) Activation of RAR-RXR heterodimers induces increased transcription of genes that possess retinoic acid response elements (RAREs; red star). (B) In the case of ATRA sufficiency in unimpaired control brains, transcription is maintained across ATRA-sensitive genes (red stars). (C) Deficiency in ATRA leads to transcriptional repression of ATRA-sensitive genes (blue star). (D) In the case of ATRA deficiency in Alzheimer’s disease brains, transcription is decreased across ATRA-sensitive genes (blue stars).
Antioxidants 12 01921 g002
Figure 3. Crosstalk between RAR/RXR and Nrf2 signaling. (A) In ATRA sufficiency, a reduced intracellular environment promotes binding of Nrf2 to KEAP1, facilitating degradation of Nrf2. In addition, Nrf2 is sequestered by RARα and RXR receptors. The availability of ATRA leads to sustained transcription of ATRA-sensitive genes, promoting neuroprotection. (B) In ATRA deficiency, a relative absence of antioxidants causes the generation of reactive oxygen species (ROS). Oxidative stress causes the liberation of Nrf2 from KEAP1, allowing Nrf2 to enter the nucleus. Within the nucleus, Nrf2 dimerizes with sMaf at antioxidant response elements (AREs), leading to increased transcription of endogenous antioxidants that serve to counteract ROS and ROS-induced damage. The relative absence of ATRA diminishes RAR/RXR-mediated signaling, leading to a downregulation of ATRA-sensitive genes that mediate neuroprotection.
Figure 3. Crosstalk between RAR/RXR and Nrf2 signaling. (A) In ATRA sufficiency, a reduced intracellular environment promotes binding of Nrf2 to KEAP1, facilitating degradation of Nrf2. In addition, Nrf2 is sequestered by RARα and RXR receptors. The availability of ATRA leads to sustained transcription of ATRA-sensitive genes, promoting neuroprotection. (B) In ATRA deficiency, a relative absence of antioxidants causes the generation of reactive oxygen species (ROS). Oxidative stress causes the liberation of Nrf2 from KEAP1, allowing Nrf2 to enter the nucleus. Within the nucleus, Nrf2 dimerizes with sMaf at antioxidant response elements (AREs), leading to increased transcription of endogenous antioxidants that serve to counteract ROS and ROS-induced damage. The relative absence of ATRA diminishes RAR/RXR-mediated signaling, leading to a downregulation of ATRA-sensitive genes that mediate neuroprotection.
Antioxidants 12 01921 g003
Figure 4. Proposed effects of age-related epigenetic changes in ATRA-sensitive and Nrf2-sensitive gene transcription, and AD progression. (A) ATRA deficiency is accompanied by a co-repressor complex that includes HDAC3. The accompanying age-related upregulation of class I/II HDACs and DMNTs further ensures transcriptional silencing of both ATRA- and Nrf2-sensitive genes. (B) The activity of ATRA as a dietary exogenous AO and transcriptionally active ligand is lost in the hippocampus, thereby increasing oxidative stress and Nrf2-mediated gene expression until epigenetic changes silence the transcription of Nrf2-driven pathways, resulting in expedited ARCD and AD progression.
Figure 4. Proposed effects of age-related epigenetic changes in ATRA-sensitive and Nrf2-sensitive gene transcription, and AD progression. (A) ATRA deficiency is accompanied by a co-repressor complex that includes HDAC3. The accompanying age-related upregulation of class I/II HDACs and DMNTs further ensures transcriptional silencing of both ATRA- and Nrf2-sensitive genes. (B) The activity of ATRA as a dietary exogenous AO and transcriptionally active ligand is lost in the hippocampus, thereby increasing oxidative stress and Nrf2-mediated gene expression until epigenetic changes silence the transcription of Nrf2-driven pathways, resulting in expedited ARCD and AD progression.
Antioxidants 12 01921 g004
Figure 5. Working model of therapeutic effects of HDAC inhibitors on the activation of RAR-, PPAR-, and Nrf2-mediated gene transcription. (A) ATRA sufficiency removes the co-repressor complex and recruits histone acetylases, histone methyltransferases, and transcription factors. Therapeutic employment of a class I/II HDAC inhibitor results in an open chromatin conformation and accessible promotor region that augments the beneficial effects of ATRA sufficiency. Also, Nrf2 is transcriptionally active in circumstances of ATRA deficiency combined with a HDAC inhibitor. (B) Sufficient hippocampal ATRA concentration staves off AD progression until age-related epigenetic changes silence ATRA- and Nrf2-sensitive gene expression. (C) With ATRA insufficiency, epigenetic intervention alone will increase compensatory Nrf2-mediated AO defenses, aiding in delaying MCI and AD, but ATRA-deficiency remains, preventing the expression of ATRA-induced neuroprotective pathways. (D) HDAC inhibition, in combination with retinoid supplementation, lifts the epigenetic silencing of RAREs, PPAREs, and AREs, and allows for the activation of both ATRA- and Nrf2-responsive neuroprotective genes.
Figure 5. Working model of therapeutic effects of HDAC inhibitors on the activation of RAR-, PPAR-, and Nrf2-mediated gene transcription. (A) ATRA sufficiency removes the co-repressor complex and recruits histone acetylases, histone methyltransferases, and transcription factors. Therapeutic employment of a class I/II HDAC inhibitor results in an open chromatin conformation and accessible promotor region that augments the beneficial effects of ATRA sufficiency. Also, Nrf2 is transcriptionally active in circumstances of ATRA deficiency combined with a HDAC inhibitor. (B) Sufficient hippocampal ATRA concentration staves off AD progression until age-related epigenetic changes silence ATRA- and Nrf2-sensitive gene expression. (C) With ATRA insufficiency, epigenetic intervention alone will increase compensatory Nrf2-mediated AO defenses, aiding in delaying MCI and AD, but ATRA-deficiency remains, preventing the expression of ATRA-induced neuroprotective pathways. (D) HDAC inhibition, in combination with retinoid supplementation, lifts the epigenetic silencing of RAREs, PPAREs, and AREs, and allows for the activation of both ATRA- and Nrf2-responsive neuroprotective genes.
Antioxidants 12 01921 g005
Table 1. Evidence for transcriptional dysregulation of ATRA transport, synthesis, and metabolism in the hippocampus of human AD. Downregulated mRNA transcripts are shown in blue, upregulated mRNA transcripts are shown in red, and transcripts that show no significant change are in shown in gray. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and differential expression (DE) score, as performed by van Rooij and colleagues [107].
Table 1. Evidence for transcriptional dysregulation of ATRA transport, synthesis, and metabolism in the hippocampus of human AD. Downregulated mRNA transcripts are shown in blue, upregulated mRNA transcripts are shown in red, and transcripts that show no significant change are in shown in gray. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and differential expression (DE) score, as performed by van Rooij and colleagues [107].
GeneFunctionlogFCp ValueFDRDE Score
RBP4Retinol transport−1.307.9 × 10−133.4 × 10−110.43
TTRRetinol transport0.571.7 × 10−12.6 × 10−10.01
STRA6Retinol neuronal import−0.726.4 × 10−21.1 × 10−10.02
RDH12Retinal synthesis−0.929.8 × 10−43.2 × 10−30.08
RBP1Retinol transport−0.783.2 × 10−94.7 × 10−80.19
ADLH1A1ATRA synthesis−0.183.2 × 10−14.2 × 10−10.00
ADLH1A2ATRA synthesis−0.471.2 × 10−11.9 × 10−10.01
ADLH1A3ATRA synthesis−0.715.0 × 10−52.3 × 10−40.09
CRABP1Nuclear translation0.383.1 × 10−14.1 × 10−10.00
CRABP2Nuclear translocation−0.419.6 × 10−21.6 × 10−10.01
FABP5Nuclear translocation−0.192.5 × 10−13.4 × 10−10.00
CYP26A1ATRA-sensitive ATRA catabolism−1.096.7 × 10−31.7 × 10−20.06
CYP26B1ATRA-sensitive ATRA catabolism−0.972.5 × 10−61.7 × 10−50.15
RARAα retinoic acid receptor−0.057.4 × 10−18.1 × 10−10.00
RARBβ retinoic acid receptor0.257.2 × 10−21.3 × 10−10.01
RARGγ retinoic acid receptor0.686.3 × 10−74.9 × 10−60.12
SP1RARG transcriptional regulator0.631.0 × 10−81.3 × 10−70.14
LRATATRA into AT-retinyl ester0.611.2 × 10−22.7 × 10−20.03
Table 2. Evidence for transcriptional block of ATRA signaling in the hippocampus of human AD. Red highlighting indicates upregulated mRNA transcripts. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and DE score, as performed by van Rooij and colleagues [107].
Table 2. Evidence for transcriptional block of ATRA signaling in the hippocampus of human AD. Red highlighting indicates upregulated mRNA transcripts. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and DE score, as performed by van Rooij and colleagues [107].
GeneFunctionlogFCp ValueFDRDE Score
NCOR1Class II nuclear receptor co-repressor0.329.5 × 10−43.1 × 10−30.03
NCOR2Class II nuclear receptor co-repressor0.102.6 × 10−13.5 × 10−10.00
ZBTB16Class II nuclear receptor co-repressor0.963.5 × 10−95.0 × 10−80.23
TNIP1RAR co-repressor0.355.9 × 10−63.6 × 10−50.05
RIF1RAR co-repressor0.232.6 × 10−25.2 × 10−20.01
LCORClass I/II nuclear receptor co-repressor0.497.7 × 10−42.6 × 10−30.04
Table 3. Indirect evidence for hippocampal ATRA depletion-induced increases in oxidative stress, inflammation, and mitochondrial dysfunction in human AD. Downregulated mRNA levels are shown in blue; upregulated mRNA levels are shown in red. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and DE score, as performed by van Rooij and colleagues [107].
Table 3. Indirect evidence for hippocampal ATRA depletion-induced increases in oxidative stress, inflammation, and mitochondrial dysfunction in human AD. Downregulated mRNA levels are shown in blue; upregulated mRNA levels are shown in red. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and DE score, as performed by van Rooij and colleagues [107].
GeneFunctionlogFCp ValueFDRDE Score
NFE2L1ROS sensor-transcription factor0.542.0 × 10−103.9 × 10−90.15
NFE2L2ROS sensor-transcription factor0.461.0 × 10−44.5 × 10−40.05
NFKB1Inflammation0.802.6 × 10−141.9 × 10−120.27
NFKB2Inflammation0.641.1 × 10−71.1 × 10−60.13
NQO1Endogenous AO−0.224.2 × 10−15.2 × 10−10.00
NQO2Endogenous AO−0.362.7 × 10−37.7 × 10−30.03
GSREndogenous AO−0.252.5 × 10−37.2 × 10−30.02
GSTA4Endogenous AO−0.927.6 × 10−161.1 × 10−130.31
GSTM4Endogenous AO−0.397.1 × 10−42.4 × 10−30.03
GSTO1Endogenous AO−0.323.4 × 10−39.2 × 10−30.02
GSTO2Endogenous AO−0.478.0 × 10−31.9 × 10−20.03
GSTZ1Endogenous AO−0.805.0 × 10−52.4 × 10−40.10
SLC7A11Cysteine/glutamate transporter−0.266.4 × 10−21.1 × 10−10.01
PTGR1Endogenous AO−0.591.3 × 10−45.3 × 10−40.06
HMOX2Endogenous AO−0.554.5 × 10−84.7 × 10−70.12
SOD1Endogenous AO−0.312.8 × 10−25.7 × 10−20.01
GLRX3Endogenous AO−0.164.7 × 10−28.7 × 10−20.01
CATEndogenous AO0.421.6 × 10−34.9 × 10−30.03
TOMM20Protein targeting to mitochondria−0.613.1 × 10−117.8 × 10−100.19
TOMM40Protein targeting to mitochondria−0.561.8 × 10−82.1 × 10−70.12
OPA1Regulates mitochondrial fusion−0.811.9 × 10−141.5 × 10−120.27
DNM1LRegulates mitochondrial fission−0.791.3 × 10−137.2 × 10−120.26
RORARetinoic acid-related orphan receptor α 0.483.2 × 10−41.2 × 10−30.05
Table 4. Indirect evidence for transcriptional dysregulation via upregulation of histone deacetylases (HDACs) in human AD. Upregulated mRNA levels are shown in red. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and DE score.
Table 4. Indirect evidence for transcriptional dysregulation via upregulation of histone deacetylases (HDACs) in human AD. Upregulated mRNA levels are shown in red. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and DE score.
GeneFunctionlogFCp ValueFDRDE Score
HDAC1Class I histone deacetylase0.508.3 × 10−88.1 × 10−70.10
HDAC2Class I histone deacetylase0.092.7 × 10−13.7 × 10−10.00
HDAC4Class II histone deacetylase0.261.2 × 10−22.8 × 10−20.01
HDAC5Class II histone deacetylase0.149.1 × 10−21.5 × 10−10.00
HDAC7Class II histone deacetylase0.661.4 × 10−97.7 × 10−30.03
Table 5. ADAM- and BACE-family members in human AD. Upregulated mRNA levels are shown in red. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and DE score.
Table 5. ADAM- and BACE-family members in human AD. Upregulated mRNA levels are shown in red. Each gene includes log fold-change (logFC), p value, false discovery rate (FDR), and DE score.
GeneFunctionlogFCp ValueFDRDE Score
ADAM10proteolytic α-secretase0.342.9 × 10−41.1 × 10−30.03
PPARαADAM10 activator0.376.7 × 10−31.7 × 10−20.02
SIRT1ADAM10 activator0.771.8 × 10−103.7 × 10−90.22
BACE1β-secretase−0.199.3 × 10−21.6 × 10−10.01
ADAM8proteolytic α-secretase0.637.5 × 10−31.8 × 10−20.04
ADAM9proteolytic α-secretase0.181.2 × 10−11.9 × 10−10.00
ADAM15proteolytic α-secretase−0.066.7 × 10−17.5 × 10−10.00
ADAM12proteolytic α-secretase0.443.9 × 10−27.5 × 10−20.02
ADAM17proteolytic α-secretase0.371.3 × 10−33.9 × 10−30.03
ADAM19proteolytic α-secretase0.377.4 × 10−21.3 × 10−10.01
ADAM33proteolytic α-secretase1.558.3 × 10−122.6 × 10−100.49
ADAM11non-proteolytic ADAM−1.365.5 × 10−97.4 × 10−80.32
BACE2neuroprotective β-secretase1.631.1 × 10−113.2 × 10−100.52
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Almaguer, J.; Hindle, A.; Lawrence, J.J. The Contribution of Hippocampal All-Trans Retinoic Acid (ATRA) Deficiency to Alzheimer’s Disease: A Narrative Overview of ATRA-Dependent Gene Expression in Post-Mortem Hippocampal Tissue. Antioxidants 2023, 12, 1921. https://doi.org/10.3390/antiox12111921

AMA Style

Almaguer J, Hindle A, Lawrence JJ. The Contribution of Hippocampal All-Trans Retinoic Acid (ATRA) Deficiency to Alzheimer’s Disease: A Narrative Overview of ATRA-Dependent Gene Expression in Post-Mortem Hippocampal Tissue. Antioxidants. 2023; 12(11):1921. https://doi.org/10.3390/antiox12111921

Chicago/Turabian Style

Almaguer, Joey, Ashly Hindle, and J. Josh Lawrence. 2023. "The Contribution of Hippocampal All-Trans Retinoic Acid (ATRA) Deficiency to Alzheimer’s Disease: A Narrative Overview of ATRA-Dependent Gene Expression in Post-Mortem Hippocampal Tissue" Antioxidants 12, no. 11: 1921. https://doi.org/10.3390/antiox12111921

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop