HDL-Mediated Cholesterol Trafficking in the Central Nervous System: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Mireia Tondo.

Alzheimer’s disease (AD) is characterized by the accumulation of extracellular amyloid beta (Aβ) and abnormally hyperphosphorylated intracellular tau filaments in neurons. Cholesterol metabolism has been extensively implicated in the pathogenesis of AD through biological, epidemiological, and genetic studies, with the APOE gene being the most reproducible genetic risk factor for the development of AD. The apolipoprotein E (ApoE) 4  genotype seems to be a disruptive element in high-density lipoprotein (HDL)-like-mediated cholesterol transport through the brain. 

  • Alzheimer’s disease
  • apolipoprotein E
  • cholesterol trafficking
  • HDL
  • central nervous sytstem
  • dementia
  • cholesterol efflux

1. Transporters of Cholesterol in the Brain

Cholesterol trafficking in the CNS involves several transporters with similar roles to those in the peripheral cells [29][1], reinforcing the relevance of the control of lipid homeostasis in the brain. Identically to peripheral tissues, cholesterol efflux in the brain takes place through aqueous diffusion facilitated by scavenger receptor class B type I (SR-BI) and by active pathways involving the ATP-binding cassette (ABC) transporters A1 and G1 (ABCA1 and ABCG1) [30][2]. In contrast to the periphery, where SR-BI is ubiquitous, in the brain, it is only expressed in astrocytes, neurons, and capillary endothelial cells [31[3][4],32], as it is regulated by the sterol regulatory element-binding protein 2 (SREBP-2) transcription factor binding sites [33,34][5][6]. In the CNS, ABC transporters have been found in neurons, astrocytes, and capillary endothelial cells, participating in protein secretion and lipidation [35,36][7][8]. Specifically, ABCA1 acts as the main cholesterol efflux regulatory protein, interacting with mildly lipidated ApoE particles to form small HDL-like lipoproteins containing phospholipids (PL) and unesterified cholesterol (UC) [29][1]. SR-BI and other transporters from the ABC family, such as ABCG1 and ABCG4, interact with already-lipidated forms [37][9], thereby completing the lipidation of small HDL-like particles and generating larger ones via the addition of more cholesterol as well as other lipids [38][10]. Once in the extracellular space, lipoproteins can be further enriched with ApoE [39,40,41][11][12][13].
ABCA1, ABCG1, and ApoE gene transcription can be modulated by the liver X receptors (LXRs) α and β, ligand-activated transcription factors that bind to DNA and form heterodimers with retinoid X receptors (RXRs) to exert their functions [42,43][14][15]. Oxysterols and 9-cis-retinoic acid (RA) are, in turn, endogenous ligands for LXRs and RXRs, respectively [44,45][16][17]. Thus, a high concentration of intracellular cholesterol or its derivatives in astrocytes activates LXR/RXR-mediated transcription for cholesterol transport proteins to facilitate efflux [46,47][18][19]. In this sense, the co-expression of ApoE with ABCA1 by the LXR/RXR system reinforces the important role of ApoE lipidation in the efflux process [48][20]. ABCA1 may also play a critical role in removing excess cholesterol from neurons, which can either be converted into cholesterol esters and kept in the cytoplasm or converted to 24S-hydroxycholesterol (HC) by 24-hydroxylase [49[21][22],50], as discussed below. 24-HC may reach the astrocytes and, through the activation of LXR, inhibit cholesterol synthesis and upregulate ABCA1, ABCG1, and ApoE levels [44][16]. Alternatively, it can be exported from the brain through the BBB [41,51,52,53][13][23][24][25]. Concerning ABCA7, this transporter is highly expressed in the brain, sharing significant homology with ABCA1 [35][7]. However, its transcription is downregulated when intracellular cholesterol concentrations are high [51][23] and upregulated through the SREBP-2 pathway when they are low [54][26]. Its role in cholesterol efflux is less known and seems to be less relevant than those of the other ABC proteins [55][27].
In addition to endogenous LXR/RXR agonists, exogenous ligands have also been described. T0901317 is a synthetic and highly selective agonist for LXRs, with a demonstrated enhancing effect on the ABCA1 and ABCG1 transporters and cholesterol efflux [42][14]. Similarly, ABCA1 activity can also be modulated by cyclic adenosine monophosphate (cAMP) via a protein kinase A (PKA)-dependent pathway. Protein kinase A increases ABCA1 gene transcription and phosphorylates ABCA1 protein, increasing its ability to export cholesterol [56][28].
Regarding cholesterol uptake, mature HDL-like particles deliver cholesterol to brain cells through the interaction of ApoE with specific lipoprotein receptors [57][29], including the low-density lipoprotein (LDL) receptor (LDLR), LDL-receptor-related protein 1 (LRP1), the very low density lipoprotein (VLDL) receptor (VLDLR), and the ApoE receptor (ApoER2), which is mainly expressed in the brain [58][30]. All of them bind ApoE and lipidated ApoE with different degrees of affinity [29,59][1][31]. Characteristically, nascent lipoproteins secreted by astrocytes show a higher affinity for LDLR, whereas CSF ApoE-containing HDL-like particles adhere strongly to LRP1 [60][32]. Despite being present in both neurons and astrocytes, LDLR is most highly expressed in glia, whereas LRP1 is more highly expressed in neurons [61,62,63,64,65][33][34][35][36][37]. Specifically, LRP1 is a critical source of cholesterol for neurite outgrowth, synaptogenesis, and remodeling; however, it may also exert negative feedback to limit the intracellular cholesterol concentration [66,67][38][39]. Regarding VLDLR and ApoER2, they are structurally very similar to the LDLR; however, they mainly bind other ligands involved in neurodevelopment and synaptic functions, such as reelin [68][40]. The information regarding cholesterol transporters and receptors potentially involved in cholesterol processes in the brain is summarized in Table 1.
Table 1.
 Cholesterol transporters and receptors potentially involved in cholesterol efflux and uptake processes in the brain.
With respect to lipid movement through the central and peripheral compartments, the BBB and the blood–cerebrospinal fluid barrier (BCSFB) act as semipermeable membranes, regulating the exchange of solutes between the blood and CNS [69][41]. Therefore, all lipoproteins, including LDL, VLDL, and chylomicrons, are excluded from the brain [70][42]. ApoA-I’s presence in the CSF derives from the blood circulating HDL [71,72][43][44]. The ABCA1 and ABCG1 transporters present on epithelial cells of the BBB can mediate the lipidation of peripheral apolipoproteins after their entry into the CNS [48[20][45],73], facilitating the formation of ApoE/ApoA-I small HDL-like particles [34][6]. Moreover, small HDL plasma particles can enter the brain via SR-BI-mediated uptake and transcytosis [34][6].
In contrast to cholesterol, the oxysterols 24-HC and 27-hydroxycholesterol (27-HC) (oxidized cholesterol metabolites) can cross the BBB and BCSFB at no energetic cost [74][46]. In that sense, 27-HC is synthesized by CYP27A1, representing the major cholesterol metabolite in the circulation. Its expression takes place in most of the organs and tissues [75][47] and can be transported by diffusion from the circulation into the brain [76][48]. Recent works support the idea that the pool of 27-HC contributes to cholesterol-related metabolite uptake in neurons [77][49]. On the other hand, due to the limited capacity of neurons and glial cell to eliminate cholesterol, spare cholesterol must reach the liver for further conversion to bile acids and final excretion. As previously stated, this function can rely on 24-HC, the main available hydrophilic form of cholesterol in the brain, to be transferred through the BBB [42,51,52,53][14][23][24][25]. 24-HC is originally released by neurons through the neuron-specific enzyme CYP46A1 [53][25]. A secondary excretion pathway through the BBB may involve cholesterol efflux mediated by ABCA1 and ABCG1 [34,78][6][50]. The steady-state levels of both oxysterols are tightly regulated in the brain. Therefore, disturbances in their concentrations have been associated with different forms of dementia [79][51]. In that sense, several works have evaluated plasma or CSF oxysterol levels in patients with AD, with evidence of impaired concentrations [80,81,82][52][53][54]. Particularly, 24-OHC has been found to regulate APP via the production of the amyloidogenic fragment [83][55], whereas 27-HC may contribute to amyloid deposition [74][46]. Higher levels of the metabolites 24-HC and 27-HC in the CSF suggest an increase in the cerebral cholesterol load, as observed in AD brains in postmortem examinations [84][56]. Overall, these results suggest a key role of these cholesterol metabolites in AD pathology. However, the relationship of HDL-like lipoproteins with these metabolites in the CNS remains largely unknown.

2. Apolipoprotein E in the CNS in the Context of AD

Excellent reviews focusing almost exclusively on ApoE and AD exist [85,86,87,88,89,90,91,92,93,94][57][58][59][60][61][62][63][64][65][66]. As previously stated, liver-synthesized ApoE cannot cross the BBB [95][67]. Thus, the ApoE in the CNS is synthesized by astrocytes and microglia [96][68]. In stressful situations, neurons can also produce ApoE [97][69]. It is worth noting that despite the fact that some authors refer to ApoE as a relevant AD component on its own, it is indeed a relevant part of the HDL-like lipoprotein structure that transports multiple proteins and lipid species, thereby affecting their CNS metabolism. Accordingly, ApoE is the main lipid carrier in the CNS, with functions that include the transport of lipids (mainly cholesterol) between neurons and glial cells via interactions with transporters such as ABCA1 and ABCG1 [98][70], the regulation of lipid metabolism [99][71], and the enhancement of axonal growth through LDLR interactions [60][32]. ApoE is also essential for the brain homeostasis-regulating processes of Aβ clearance as well as for the inhibition of inflammatory pathways, both functions of great importance in the AD brain, where Aβ oligomers and neuroinflammatory metabolites tend to accumulate [100][72]. Specifically, lipidated ApoE clears Aβ peptides from the brain [101][73]. In that sense, it is a well-established fact that the ApoE lipidation degree is positively associated with its affinity for soluble Aβ, with poorly lipidated ApoE obstructing Aβ clearance and stimulating Aβ deposition [25[74][75][76][77],102,103,104], thus representing a risk factor for AD.

3. HDL-like Lipoprotein Metabolism in the CNS

To understand the HDL-mediated cholesterol efflux and uptake processes through CNS cells in AD, a brief explanation of the biogenesis, remodeling, and delivery of HDL-like particles is provided below. Like plasma, the lipoproteins present in the CSF are composed of cholesterol, phospholipids, and apolipoproteins. Overall, their brain concentration is very low, representing approximately 1–10% of their plasma levels [132][78]. Brain lipoproteins present with a similar size and density to peripheral HDL particles, and therefore they are defined as “HDL-like particles” [132][78]. However, brain lipoproteins present with unique traits, including a wider size range (8–22 nm) and a different apolipoprotein composition, with ApoE being the major protein component [99][71]. In addition to ApoE, glial-derived discoidal HDL has ApoJ, whereas mature spherical CSF HDL has small amounts of ApoA-I. As stated above, ApoA-I is the most important constituent of plasma HDL; however, it is not produced in the brain and is, rather, delivered to the CNS through the BBB [133][79]. Other minoritarian apolipoproteins present in the brain include ApoA-II, ApoA-IV, ApoD, and ApoH [132][78]. Despite not being fully elucidated, the mechanisms involved in CSF lipoprotein synthesis and remodeling are similar to those observed for plasma HDL. It is well-known that astrocytes and microglia are responsible for the synthesis of most lipoproteins found in the brain and CSF [134][80]. Remodeling enzymes and lipid transfer proteins have also been identified in the CNS [135][81]. Lecithin-cholesterol acyltransferase (LCAT) converts UC and phosphatidylcholine to cholesteryl esters [136][82] via ApoE activation [137][83]. In fact, LCAT can be synthesized in the liver and testes as well as in astrocytes [137][83], suggesting its important role in the remodeling and maturation of nascent HDL-like proteins into larger spherical particles [99][71]. It has been found at concentrations representing around 5% of the levels in plasma [99,138][71][84]. Other remodeling enzymes include phospholipid transfer protein (PLTP), which is reported to be at a concentration representing about 15% of the plasma level [139[85][86],140], and cholesteryl ester transfer protein (CETP), which is reported to be present at about 12% of the level found in plasma [140][86] or non-existent [141][87]. Finally, as described above, cholesterol can be delivered to neurons by mature HDL-like lipoproteins through interactions with specific receptors [8][88]. A detailed description of HDL-like-mediated cholesterol trafficking in healthy and AD brains is shown in Figure 1.
Figure 1. Schematic representation of the main steps involved in cholesterol trafficking in the brain. (A) In healthy subjects, astrocytes are responsible for de novo cholesterol and ApoE synthesis, with ApoE3 being the predominant isoform. Cholesterol efflux from astrocytes occurs, in part, through the ABCA1 and ABCG1 transporters. Lipid-free ApoE and, in smaller amounts, ApoA-I and ApoJ can be further lipidized by remodeling enzymes, resulting in spherical mature HDL-like particles that can interact with membrane receptors such as LRP1 and LDLR, leading to cholesterol uptake by neurons and guaranteeing essential functions such as synaptogenesis and neurite growth. Oxysterols can flux across the BBB. Neurons convert excess cholesterol in 24-HC, which can be eliminated to the bloodstream. In contrast, 27-HC enters the brain, where it promotes various functions. ApoE also contributes to the clearance of Aβ peptides. (B) In AD subjects, the pathological accumulation of hyperphosphorylated tau protein and Aβ plaque deposition may alter physiological functions in the brain. ApoE4, the predominant isoform in AD patients, is poorly lipidated and barely removes Aβ peptides. LRP1 plays a critical role in neuronal tau endocytosis. Recent works suggest alterations regarding cholesterol transport, including reduced HDL-like-mediated cholesterol efflux and impaired cholesterol uptake, leading to cell dysfunction. ABC: ATP-binding cassette; AD: Alzheimer’s disease; Apo: apolipoprotein; Aβ: amyloid beta; BBB: blood–brain barrier; CETP: cholesteryl ester transfer protein; HC: hydroxycholesterol; HDL: high-density lipoprotein; LCAT: lecithin-cholesterol acyltransferase; LDLR: low-density lipoprotein receptor; LRP1: LDLR-related protein 1.

4. Cholesterol Efflux to HDL-like Particles in AD

Brain cholesterol efflux is a key step in cholesterol trafficking through the CNS. As previously described, the process consists of the transference of intracellular cholesterol to extracellular acceptors, including apolipoproteins and HDL-like lipoproteins, through aqueous diffusion or SR-BI and ABC-family transporters. The first study that aimed to describe the ability of brain lipoproteins to promote cholesterol efflux was performed in 1998 in fibroblasts [142][89]. This study was pioneering in demonstrating the critical role of CSF lipoproteins in brain cholesterol trafficking, thus opening the door for further studies. Since then, researchers have explored the ability of cholesterol acceptors, including serum, serum-isolated, or recombinant apolipoproteins, lipoprotein, and CSF, to promote cholesterol efflux in different cell types. Considering the effect of ApoE isoforms on AD pathology, some works evaluated how they affect efflux capacity and obtained divergent results. While some authors found similar rates of cholesterol efflux from HDL-like particles in neuronal cells and astrocytes regardless of the ApoE isoform [145[90][91],146], others demonstrated that recombinant ApoE3 was able to induce a higher lipid efflux compared to recombinant ApoE4 in neuronal and astrocytic cell cultures [147][92]. Accordingly, another work also found that primary astrocytes from modified mice expressing human ApoE4 displayed lower cholesterol efflux than those cells from mice expressing human ApoE3 [110][93]. Interestingly, the accumulation of fat in astrocytes, a stress-associated condition, induced the assembly and secretion of potentially toxic triacylglycerol-rich ApoE-containing lipoproteins, a process enhanced by ApoE4 [146][91]. On the whole, the contribution of the ApoE genotype to cholesterol efflux and its pathophysiological consequences requires further studies. Finally, another line of work has focused on exploring the cholesterol efflux capacity of human samples taken from control individuals and AD patients. Several studies using cultured murine macrophages found reduced cholesterol efflux in AD patients’ samples when using serum as an acceptor [30,148][2][94]. Despite these interesting results, some authors argued that CSF is the most suitable physiological sample to study cholesterol trafficking in the brain because it contains all the components involved in central cholesterol efflux. Accordingly, the first approach assessing cholesterol efflux capacity in a small number of control and AD patient CSF samples in rat astrocytes found no differences between the two groups [141][87]. More recently, the evaluation of cholesterol efflux to the CSF in larger cohorts was addressed. Using baby hamster kidney (BHK) cells overexpressing ABCA1, the authors reported that CSF samples taken from AD patients displayed 30% less cholesterol efflux than CSF samples from cognitively healthy participants [149][95]. In agreement, a recent work performed with murine macrophages induced with cAMP to express ABCA1 at one end and Chinese hamster ovary (CHO)-K1 cells overexpressing ABCG1 at the other end resulted in decreased levels of cholesterol efflux from AD patients’ CSF compared to a control population [150][96]. Likewise, cholesterol efflux to the CSF from ApoE3 carriers was significantly increased compared to that from ApoE4 carriers in ABCA1-induced BHK cells [110][93]. Despite the importance of these findings, only one work has performed similar studies in cells that are functionally and morphologically relevant in the brain and, thus, for AD pathology [151][97], representing a proof of concept for CSF cholesterol efflux quantification in human neurological cells. Efflux measurement in neurons, microglia, and astrocytes allowed researchers to conclude that CSF cholesterol efflux levels are positively correlated with total cholesterol, ApoA-I, ApoE, and ApoJ concentrations. However, demographic and clinical data were unknown because the CSF samples were taken from anonymous patients, making it impossible to compare efflux among healthy controls and patients with AD [151][97].

5. Cholesterol HDL-like Uptake in AD

One of the first studies addressing cholesterol uptake in connection with AD was performed in 1998 [142][89]. Specifically, the work addressed the function of CSF lipoproteins with assays of cholesterol efflux and cholesterol uptake. The authors showed that CSF lipoproteins labeled with a fluorescent dye were internalized by neuroglioma cells and cultured primary neurons and astrocytes. Nonetheless, this field has scarcely been explored in later years because most studies focused almost exclusively on cholesterol efflux. A recent report demonstrated that astrocytic ApoE-derived lipoproteins, presumably HDL-like, can transport a variety of microRNAs that specifically downregulate the genes involved in neuronal cholesterol biosynthesis [152][98]. Importantly, these ApoE-induced effects were significantly abolished in neurons after silencing LDLR or LRP1. Furthermore, ApoE4 was less capable of regulating these neuronal pathways in this situation [152][98]. An important line of work regarding cholesterol uptake involves the proprotein convertase subtilisin/kexin type 9 (PCSK9), a protein whose main role is to regulate LDLR recycling. PCSK9 is present in the serum and CSF. In the periphery, PCSK9 degrades LDLR, while in neurons it degrades LDLR as well as other ApoE-binding receptors, including VLDLR, LRP1, and apoER2 [89][61], causing decreased cholesterol uptake. Interestingly, recent studies found that patients with AD presented with significantly increased PCSK9 concentrations [153[99][100],154], especially ApoE4 carrier patients [153][99]. These results involve cholesterol uptake as a potential pathological mechanism of AD. It should be noted that LRP1 is a critical receptor that mediates tau endocytosis and spread [129][101].

References

  1. Mahley, R.W. Central Nervous System Lipoproteins: ApoE and Regulation of Cholesterol Metabolism. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 1305–1315.
  2. Camponova, P.; le Page, A.; Berrougui, H.; Lamoureux, J.; Pawelec, G.; Witkowski, M.J.; Fulop, T.; Khalil, A.; Slimane, M.; Mellal, B. Alteration of High-Density Lipoproteins Functionality in Alzheimer’s Disease Patients. Can. J. Physiol. Pharmacol. 2017, 95, 894–903.
  3. Sovic, A.; Balazs, Z.; Hrzenjak, A.; Reicher, H.; Panzenboeck, U.; Malle, E.; Sattler, W. Scavenger Receptor Class B, Type I Mediates Uptake of Lipoprotein-Associated Phosphatidylcholine by Primary Porcine Cerebrovascular Endothelial Cells. Neurosci. Lett. 2004, 368, 11–14.
  4. Husemann, J.; Loike, J.D.; Kodama, T.; Silverstein, S.C. Scavenger Receptor Class B Type I (SR-BI) Mediates Adhesion of Neonatal Murine Microglia to Fibrillar β-Amyloid. J. Neuroimmunol. 2001, 114, 142–150.
  5. Khalil, A.; Berrougui, H.; Pawelec, G.; Fulop, T. Impairment of the ABCA1 and SR-BI-Mediated Cholesterol Efflux Pathways and HDL Anti-Inflammatory Activity in Alzheimer’s Disease. Mech. Ageing Dev. 2012, 133, 20–29.
  6. Panzenboeck, U.; Balazs, Z.; Sovic, A.; Hrzenjak, A.; Levak-Frank, S.; Wintersperger, A.; Malle, E.; Sattler, W. ABCA1 and Scavenger Receptor Class B, Type I, Are Modulators of Reverse Sterol Transport at an in Vitro Blood-Brain Barrier Constituted of Porcine Brain Capillary Endothelial Cells. J. Biol. Chem. 2002, 277, 42781–42789.
  7. Kim, W.S.; Guillemin, G.J.; Glaros, E.N.; Lim, C.K.; Garner, B. Quantitation of ATP-Binding Cassette Subfamily-A Transporter Gene Expression in Primary Human Brain Cells. NeuroReport 2006, 17, 891–896.
  8. Tarr, P.T.; Edwards, P.A. ABCG1 and ABCG4 are Coexpressed in Neurons and Astrocytes of the CNS and Regulate Cholesterol Homeostasis through SREBP-2. J. Lipid Res. 2008, 49, 169–182.
  9. Ben Aissa, M.; Lewandowski, C.T.; Ratia, K.M.; Lee, S.H.; Layden, B.T.; Ladu, M.J.; Thatcher, G.R.J. Discovery of Nonlipogenic ABCA1 Inducing Compounds with Potential in Alzheimer’s Disease and Type 2 Diabetes. ACS Pharmacol. Transl. Sci. 2021, 4, 143–154.
  10. Vaughan, A.M.; Oram, J.F. ABCA1 and ABCG1 or ABCG4 Act Sequentially to Remove Cellular Cholesterol and Generate Cholesterol-Rich HDL. J. Lipid Res. 2006, 47, 2433–2443.
  11. Hirsch-Reinshagen, V.; Zhou, S.; Burgess, B.L.; Bernier, L.; McIsaac, S.A.; Chan, J.Y.; Tansley, G.H.; Cohn, J.S.; Hayden, M.R.; Wellington, C.L. Deficiency of ABCA1 Impairs Apolipoprotein E Metabolism in Brain. J. Biol. Chem. 2004, 279, 41197–41207.
  12. Pitas, R.E.; Boyles, J.K.; Lee, S.H.; Foss, D.; Mahley, R.W. Astrocytes Synthesize Apolipoprotein E and Metabolize Apolipoprotein E-Containing Lipoproteins. Biochim. Biophys. Acta (BBA)—Lipids Lipid Metab. 1987, 917, 148–161.
  13. Wahrle, S.E.; Jiang, H.; Parsadanian, M.; Legleiter, J.; Han, X.; Fryer, J.D.; Kowalewski, T.; Holtzman, D.M. ABCA1 Is Required for Normal Central Nervous System ApoE Levels and for Lipidation of Astrocyte-Secreted ApoE. J. Biol. Chem. 2004, 279, 40987–40993.
  14. Eckert, G.P.; Vardanian, L.; Rebeck, G.W.; Burns, M.P. Regulation of Central Nervous System Cholesterol Homeostasis by the Liver X Receptor Agonist TO-901317. Neurosci. Lett. 2007, 423, 47–52.
  15. Fukumoto, H.; Deng, A.; Irizarry, M.C.; Fitzgerald, M.L.; Rebeck, G.W. Induction of the Cholesterol Transporter ABCA1 in Central Nervous System Cells by Liver X Receptor Agonists Increases Secreted Aβ Levels. J. Biol. Chem. 2002, 277, 48508–48513.
  16. Abildayeva, K.; Jansen, P.J.; Hirsch-Reinshagen, V.; Bloks, V.W.; Bakker, A.H.F.; Ramaekers, F.C.S.; de Vente, J.; Groen, A.K.; Wellington, C.L.; Kuipers, F.; et al. 24(S)-Hydroxycholesterol Participates in a Liver X Receptor-Controlled Pathway in Astrocytes That Regulates Apolipoprotein E-Mediated Cholesterol Efflux. J. Biol. Chem. 2006, 281, 12799–12808.
  17. Koldamova, R.P.; Lefterov, I.M.; Ikonomovic, M.D.; Skoko, J.; Lefterov, P.I.; Isanski, B.A.; DeKosky, S.T.; Lazo, J.S. 22R-Hydroxycholesterol and 9-Cis-Retinoic Acid Induce ATP-Binding Cassette Transporter A1 Expression and Cholesterol Efflux in Brain Cells and Decrease Amyloid β Secretion. J. Biol. Chem. 2003, 278, 13244–13256.
  18. Benarroch, E.E. Brain Cholesterol Metabolism and Neurologic Disease. Neurology 2008, 71, 1368–1373.
  19. Jeong, W.; Lee, H.; Cho, S.; Seo, J. ApoE4-Induced Cholesterol Dysregulation and Its Brain Cell Type-Specific Implications in the Pathogenesis of Alzheimer’s Disease. Mol. Cells 2019, 42, 739.
  20. Martinez, A.E.; Weissberger, G.; Kuklenyik, Z.; He, X.; Meuret, C.; Parekh, T.; Rees, J.C.; Parks, B.A.; Gardner, M.S.; King, S.M. The Small HDL Particle Hypothesis of Alzheimer’s Disease. Alzheimer’s Dement. 2022, 1–14.
  21. Lund, E.G.; Xie, C.; Kotti, T.; Turley, S.D.; Dietschy, J.M.; Russell, D.W. Knockout of the Cholesterol 24-Hydroxylase Gene in Mice Reveals a Brain-Specific Mechanism of Cholesterol Turnover. J. Biol. Chem. 2003, 278, 22980–22988.
  22. Lütjohann, D.; Breuer, O.; Ahlborg, G.; Nennesmo, I.; Sidén, Å.; Diczfalusy, U.; Björkhem, I. Cholesterol Homeostasis in Human Brain: Evidence for an Age-Dependent Flux of 24S-Hydroxycholesterol from the Brain into the Circulation. Proc. Natl. Acad. Sci. USA 1996, 93, 9799.
  23. Chang, T.Y.; Yamauchi, Y.; Hasan, M.T.; Chang, C. Cellular Cholesterol Homeostasis and Alzheimer’s Disease. J. Lipid Res. 2017, 58, 2239–2254.
  24. Chen, J.; Zhang, X.; Kusumo, H.; Costa, L.G.; Guizzetti, M. Cholesterol Efflux Is Differentially Regulated in Neurons and Astrocytes: Implications for Brain Cholesterol Homeostasis. Biochim. Biophys. Acta—Mol. Cell Biol. Lipids 2013, 1831, 263–275.
  25. Feringa, F.M.; van der Kant, R. Cholesterol and Alzheimer’s Disease; From Risk Genes to Pathological Effects. Front. Aging Neurosci. 2021, 13, 333.
  26. Iwamoto, N.; Abe-Dohmae, S.; Sato, R.; Yokoyama, S. ABCA7 Expression Is Regulated by Cellular Cholesterol through the SREBP2 Pathway and Associated with Phagocytosis. J. Lipid Res. 2006, 47, 1915–1927.
  27. Tomioka, M.; Toda, Y.; Mañucat, N.B.; Akatsu, H.; Fukumoto, M.; Kono, N.; Arai, H.; Kioka, N.; Ueda, K. Lysophosphatidylcholine Export by Human ABCA7. Biochim. Biophys. Acta (BBA)—Mol. Cell Biol. Lipids 2017, 1862, 658–665.
  28. Mulay, V.; Wood, P.; Rentero, C.; Enrich, C.; Grewal, T. Signal Transduction Pathways Provide Opportunities to Enhance HDL and ApoAI-Dependent Reverse Cholesterol Transport. Curr. Pharm. Biotechnol. 2012, 13, 352–364.
  29. Herz, J. ApoE Receptors in the Nervous System. Curr. Opin. Lipidol. 2009, 20, 190.
  30. Bassett, C.N.; Montine, K.S.; Neely, M.D.; Swift, L.L.; Montine, T.J. Cerebrospinal Fluid Lipoproteins in Alzheimer’s Disease. J. Neurosci. Res. 2000, 50, 282–286.
  31. Turri, M.; Marchi, C.; Adorni, M.P.; Calabresi, L.; Zimetti, F. Emerging Role of HDL in Brain Cholesterol Metabolism and Neurodegenerative Disorders. Biochim. Biophys. Acta (BBA)—Mol. Cell Biol. Lipids 2022, 1867, 159123.
  32. Holtzman, D.M.; Herz, J.; Bu, G. Apolipoprotein E and Apolipoprotein E Receptors: Normal Biology and Roles in Alzheimer’s Disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006312.
  33. Ishiguro, M.; Imai, Y.; Kohsaka, S. Expression and Distribution of Low Density Lipoprotein Receptor-Related Protein MRNA in the Rat Central Nervous System. Mol. Brain Res. 1995, 33, 37–46.
  34. Rapp, A.; Gmeiner, B.; Hüttinger, M. Implication of ApoE Isoforms in Cholesterol Metabolism by Primary Rat Hippocampal Neurons and Astrocytes. Biochimie 2006, 88, 473–483.
  35. Swanson, L.W.; Simmons, D.M.; Hofmannt, S.L.; Goldsteint, J.L.; Brownt, M.S. Localization of MRNA for Low Density Lipoprotein Receptor and a Cholesterol Synthetic Enzyme in Rabbit Nervous System by in Situ Hybridization (3-Hydroxy-3-Methylglutaryl-Coenzyme A Synthase/Apoprotein E/Myelination/Watanabe Heritable-Hyperlipidemic Rabbit). Proc. Natl. Acad. Sci. USA 1988, 85, 9821–9825.
  36. Tooyama, I.; Kawamata, T.; Akiyama, H.; Moestrup, S.K.; Gliemann, J.; McGeer, P.L. Immunohistochemical Study of A2 Macroglobulin Receptor in Alzheimer and Control Postmortem Human Brain. Mol. Chem. Neuropathol. 1993, 18, 153–160.
  37. Tooyama, I.; Kawamata, T.; Akiyama, H.; Kimura, H.; Moestrup, S.K.; Gliemann, J.; Matsuo, A.; McGeer, P.L. Subcellular Localization of the Low Density Lipoprotein Receptor-Related Protein (A2-Macroglobulin Receptor) in Human Brain. Brain Res. 1995, 691, 235–238.
  38. Lewandowski, C.T.; Laham, M.S.; Thatcher, G.R.J. Remembering Your A, B, C’s: Alzheimer’s Disease and ABCA1. Acta Pharm. Sin. B 2022, 12, 995–1018.
  39. El Asmar, Z.; Terrand, J.; Jenty, M.; Host, L.; Mlih, M.; Zerr, A.; Justiniano, H.; Matz, R.L.; Boudier, C.; Scholler, E.; et al. Convergent Signaling Pathways Controlled by LRP1 (Receptor-Related Protein 1) Cytoplasmic and Extracellular Domains Limit Cellular Cholesterol Accumulation. J. Biol. Chem. 2016, 291, 5116.
  40. Lee, G.H.; D’Arcangelo, G. New Insights into Reelin-Mediated Signaling Pathways. Front. Cell. Neurosci. 2016, 10, 122.
  41. Abbott, N.J.; Patabendige, A.A.K.; Dolman, D.E.M.; Yusof, S.R.; Begley, D.J. Structure and Function of the Blood–Brain Barrier. Neurobiol. Dis. 2010, 37, 13–25.
  42. Bahrami, A.; Barreto, G.E.; Lombardi, G.; Pirro, M.; Sahebkar, A. Emerging Roles for High-Density Lipoproteins in Neurodegenerative Disorders. Biofactors 2019, 45, 725–739.
  43. Balazs, Z.; Panzenboeck, U.; Hammer, A.; Sovic, A.; Quehenberger, O.; Malle, E.; Sattler, W. Uptake and Transport of High-Density Lipoprotein (HDL) and HDL-Associated α-Tocopherol by an in Vitro Blood–Brain Barrier Model. J. Neurochem. 2004, 89, 939–950.
  44. Zhou, A.L.; Swaminathan, S.K.; Curran, G.L.; Poduslo, J.F.; Lowe, V.J.; Li, L.; Kandimalla, K.K. Apolipoprotein A-I Crosses the Blood-Brain Barrier through Clathrin-Independent and Cholesterol-Mediated Endocytosis. J. Pharmacol. Exp. Ther. 2019, 369, 481.
  45. Kober, A.C.; Manavalan, A.P.C.; Tam-Amersdorfer, C.; Holmér, A.; Saeed, A.; Fanaee-Danesh, E.; Zandl, M.; Albrecher, N.M.; Björkhem, I.; Kostner, G.M.; et al. Implications of Cerebrovascular ATP-Binding Cassette Transporter G1 (ABCG1) and Apolipoprotein M in Cholesterol Transport at the Blood-Brain Barrier. Biochim. Biophys. Acta—Mol. Cell Biol. Lipids 2017, 1862, 573–588.
  46. Bjorkhem, I.; Heverin, M.; Leoni, V.; Meaney, S.; Diczfalusy, U. Oxysterols and Alzheimer’s Disease. Acta Neurol. Scand. 2006, 114, 43–49.
  47. Staurenghi, E.; Giannelli, S.; Testa, G.; Sottero, B.; Leonarduzzi, G.; Gamba, P. Cholesterol Dysmetabolism in Alzheimer’s Disease: A Starring Role for Astrocytes? Antioxidants 2021, 10, 1890.
  48. Heverin, M.; Meaney, S.; Lütjohann, D.; Diczfalusy, U.; Wahren, J.; Björkhem, I. Crossing the Barrier: Net Flux of 27-Hydroxycholesterol into the Human Brain. J. Lipid Res. 2005, 46, 1047–1052.
  49. Wang, Y.; Zhang, X.; Wang, T.; Liu, W.; Wang, L.; Hao, L.; Ju, M.; Xiao, R. 27-Hydroxycholesterol Promotes the Transfer of Astrocyte-Derived Cholesterol to Neurons in Co-Cultured SH-SY5Y Cells and C6 Cells. Front. Cell Dev. Biol. 2020, 8, 580599.
  50. Dietschy, J.M. Central Nervous System: Cholesterol Turnover, Brain Development and Neurodegeneration. Biol. Chem. 2009, 390, 287–293.
  51. Rantham Prabhakara, J.P.; Feist, G.; Thomasson, S.; Thompson, A.; Schommer, E.; Ghribi, O. Differential Effects of 24-Hydroxycholesterol and 27-Hydroxycholesterol on Tyrosine Hydroxylase and α-Synuclein in Human Neuroblastoma SH-SY5Y Cells. J. Neurochem. 2008, 107, 1722–1729.
  52. Leoni, V. Oxysterols as Markers of Neurological Disease—A Review. Scand. J. Clin. Lab. Investig. 2009, 69, 22–25.
  53. Hughes, T.M.; Rosano, C.; Evans, R.W.; Kuller, L.H. Brain Cholesterol Metabolism, Oxysterols, and Dementia. J. Alzheimers Dis. 2013, 33, 891.
  54. Leoni, V.; Solomon, A.; Lövgren-Sandblom, A.; Minthon, L.; Blennow, K.; Hansson, O.; Wahlund, L.O.; Kivipelto, M.; Björkhem, I. Diagnostic Power of 24S-Hydroxycholesterol in Cerebrospinal Fluid: Candidate Marker of Brain Health. J. Alzheimer’s Dis. 2013, 36, 739–747.
  55. Djelti, F.; Braudeau, J.; Hudry, E.; Dhenain, M.; Varin, J.; Bièche, I.; Marquer, C.; Chali, F.; Ayciriex, S.; Auzeil, N.; et al. CYP46A1 Inhibition, Brain Cholesterol Accumulation and Neurodegeneration Pave the Way for Alzheimer’s Disease. Brain 2015, 138, 2383–2398.
  56. Fonseca, A.C.R.G.; Resende, R.; Oliveira, C.R.; Pereira, C.M.F. Cholesterol and Statins in Alzheimer’s Disease: Current Controversies. Exp. Neurol. 2010, 223, 282–293.
  57. Fernandez, C.G.; Hamby, M.E.; McReynolds, M.L.; Ray, W.J. The Role of APOE4 in Disrupting the Homeostatic Functions of Astrocytes and Microglia in Aging and Alzheimer’s Disease. Front. Aging Neurosci. 2019, 11, 14.
  58. Marsillach, J.; Adorni, M.P.; Zimetti, F.; Papotti, B.; Zuliani, G.; Cervellati, C. HDL Proteome and Alzheimer’s Disease: Evidence of a Link. Antioxidants 2020, 9, 1224.
  59. Van Valkenburgh, J.; Meuret, C.; Martinez, A.E.; Kodancha, V.; Solomon, V.; Chen, K.; Yassine, H.N. Understanding the Exchange of Systemic HDL Particles Into the Brain and Vascular Cells Has Diagnostic and Therapeutic Implications for Neurodegenerative Diseases. Front. Physiol. 2021, 12, 1335.
  60. Shobab, L.A.; Hsiung, G.Y.R.; Feldman, H.H. Cholesterol in Alzheimer’s Disease. Lancet Neurol. 2005, 4, 841–852.
  61. Poirier, J. Apolipoprotein E Represents a Potent Gene-Based Therapeutic Target for the Treatment of Sporadic Alzheimer’s Disease. Alzheimer’s Dement. 2008, 4, S91–S97.
  62. Hirsch-Reinshagen, V.; Wellington, C.L. Cholesterol Metabolism, Apolipoprotein E, Adenosine Triphosphate-Binding Cassette Transporters, and Alzheimer’s Disease. Curr. Opin. Lipidol. 2007, 18, 325–332.
  63. Poirier, J. Apolipoprotein E, Cholesterol Transport and Synthesis in Sporadic Alzheimer’s Disease. Neurobiol. Aging 2005, 26, 355–361.
  64. Mahley, R.W.; Weisgraber, K.H.; Huang, Y. INAUGURAL ARTICLE by a Recently Elected Academy Member:Apolipoprotein E4: A Causative Factor and Therapeutic Target in Neuropathology, Including Alzheimer’s Disease. Proc. Natl. Acad. Sci. USA 2006, 103, 5644.
  65. Mahley, R.; Huang, Y.; Weisgraber, K. Detrimental Effects of Apolipoprotein E4: Potential Therapeutic Targets in Alzheimers Disease. Curr. Alzheimer Res. 2007, 4, 537–540.
  66. Fenili, D.; McLaurin, J.A. Cholesterol and Apoe: A Target for Alzheimer’s Disease Therapeutics. Curr. Drug Targets CNS Neurol. Disord. 2005, 4, 553–567.
  67. Elliott, D.A.; Weickert, C.S.; Garner, B. Apolipoproteins in the Brain: Implications for Neurological and Psychiatric Disorders. Clin. Lipidol. 2017, 5, 555–573.
  68. Yamasaki, R.; Lu, H.; Butovsky, O.; Ohno, N.; Rietsch, A.M.; Cialic, R.; Wu, P.M.; Doykan, C.E.; Lin, J.; Cotleur, A.C.; et al. Differential Roles of Microglia and Monocytes in the Inflamed Central Nervous System. J. Exp. Med. 2014, 211, 1533–1549.
  69. Frisoni, G.B.; Geroldi, C.; Blanchettl, A.; Trabucch, M.I.; Govoni, S.; Franceschini, G.; Calabresi, L. Apolipoprotein E Epsilon 4 Allele Frequency in Vascular Dementia and Alzheimer’s Disease. Stroke 1994, 25, 1703.
  70. MS Hartz, A.; Bauer, B.A.B.C. ABC Transporters in the CNS—An Inventory. Curr. Pharm. Biotechnol. 2011, 12, 656–673.
  71. Vance, J.E.; Hayashi, H. Formation and Function of Apolipoprotein E-Containing Lipoproteins in the Nervous System. Biochim. Biophys. Acta (BBA)—Mol. Cell Biol. Lipids 2010, 1801, 806–818.
  72. Flowers, S.A.; Rebeck, G.W. APOE in the Normal Brain. Neurobiol. Dis. 2020, 136, 104724.
  73. Robert, J.; Button, E.B.; Yuen, B.; Gilmour, M.; Kang, K.; Bahrabadi, A.; Stukas, S.; Zhao, W.; Kulic, I.; Wellington, C.L. Clearance of Beta-Amyloid Is Facilitated by Apolipoprotein E and Circulating High-Density Lipoproteins in Bioengineered Human Vessels. eLife 2017, 6, e29595.
  74. Strittmatter, W.J.; Weisgraber, K.H.; Huang, D.Y.; Dong, L.M.; Salvesen, G.S.; Pericak-Vance, M.; Schmechel, D.; Saunders, A.M.; Goldgaber, D.; Roses, A.D. Binding of Human Apolipoprotein E to Synthetic Amyloid Beta Peptide: Isoform-Specific Effects and Implications for Late-Onset Alzheimer Disease. Proc. Natl. Acad. Sci. USA 1993, 90, 8098–8102.
  75. Ma, J.; Yee, A.; Brewer, H.B.; Das, S.; Potter, H. Amyloid-Associated Proteins A1-Antichymotrypsin and Apolipoprotein E Promote Assembly of Alzheimer β-Protein into Filaments. Nature 1994, 372, 92–94.
  76. Sanan, D.A.; Weisgraber, K.H.; Russell, S.J.; Mahley, R.W.; Huang, D.; Saunders, A.; Schmechel, D.; Wisniewski, T.; Frangione, B.; Roses, A.D.; et al. Apolipoprotein E Associates with Beta Amyloid Peptide of Alzheimer’s Disease to Form Novel Monofibrils. Isoform ApoE4 Associates More Efficiently than ApoE3. J. Clin. Investig. 1994, 94, 860–869.
  77. Liao, H.; Zhu, Z.; Peng, Y. Potential Utility of Retinal Imaging for Alzheimer’s Disease: A Review. Front. Aging Neurosci. 2018, 10, 188.
  78. Vitali, C.; Wellington, C.L.; Calabresi, L. HDL and Cholesterol Handling in the Brain. Cardiovasc. Res. 2014, 103, 405–413.
  79. Koch, S.; Donarski, N.; Goetze, K.; Kreckel, M.; Stuerenburg, H.-J.; Buhmann, C.; Beisiegel, U. Characterization of Four Lipoprotein Classes in Human Cerebrospinal Fluid. J. Lipid Res. 2001, 42, 1143–1151.
  80. Ladu, M.J.; Reardon, C.; van Eldik, L.; Fagan, A.M.; Bu, G.; Holtzman, D.; Getz, G.S. Lipoproteins in the Central Nervous System. Ann. N.Y. Acad. Sci. 2000, 903, 167–175.
  81. Yu, C.; Youmans, K.L.; LaDu, M.J. Lipoprotein Remodelling in the Periphery: A Model for the Brain? Biochim. Biophys. Acta 2010, 1801, 819.
  82. Dobiášová, M.; Frohlich, J.J. Advances in Understanding of the Role of Lecithin Cholesterol Acyltransferase (LCAT) in Cholesterol Transport. Clin. Chim. Acta 1999, 286, 257–271.
  83. Hirsch-Reinshagen, V.; Donkin, J.; Stukas, S.; Chan, J.; Wilkinson, A.; Fan, J.; Parks, J.S.; Kuivenhoven, J.A.; Lütjohann, D.; Pritchard, H.; et al. LCAT Synthesized by Primary Astrocytes Esterifies Cholesterol on Glia-Derived Lipoproteins. J. Lipid Res. 2009, 50, 885–893.
  84. La Marca, V.; Maresca, B.; Spagnuolo, M.S.; Cigliano, L.; Dal Piaz, F.; di Iorio, G.; Abrescia, P. Lecithin-Cholesterol Acyltransferase in Brain: Does Oxidative Stress Influence the 24-Hydroxycholesterol Esterification? Neurosci. Res. 2016, 105, 19–27.
  85. Manavalan, A.P.C.; Kober, A.; Metso, J.; Lang, I.; Becker, T.; Hasslitzer, K.; Zandl, M.; Fanaee-Danesh, E.; Brijesh Pippal, J.; Sachdev, V.; et al. Phospholipid Transfer Protein Is Expressed in Cerebrovascular Endothelial Cells and Involved in High Density Lipoprotein Biogenesis and Remodeling at the Blood-Brain Barrier. J. Biol. Chem. 2014, 289, 4683–4698.
  86. Albers, J.J.; Tollefson, J.H.; Wolfbauer, G.; Albright, R.E. Cholesteryl Ester Transfer Protein in Human Brain. Int. J. Clin. Lab. Res. 1992, 21, 264–266.
  87. Demeester, N.; Castro, G.; Desrumaux, C.; de Geitere, C.; Fruchart, J.; Santens, P.; Mulleners, E.; Engelborghs, S.; de Deyn, P.; Vandekerckhove, J.; et al. Characterization and Functional Studies of Lipoproteins, Lipid Transfer Proteins, and Lecithin:Cholesterol Acyltransferase in CSF of Normal Individuals and Patients with Alzheimer’s Disease. J. Lipid Res. 2000, 84, 276–294.
  88. Zhang, J.; Liu, Q. Cholesterol Metabolism and Homeostasis in the Brain. Protein Cell 2015, 6, 254–264.
  89. Rebeck, G.W.; Alonzo, N.C.; Berezovska, O.; Harr, S.D.; Knowles, R.B.; Growdon, J.H.; Hyman, B.T.; Mendez, A.J. Structure and Functions of Human Cerebrospinal Fluid Lipoproteins from Individuals of Different APOE Genotypes. Exp. Neurol. 1998, 149, 175–182.
  90. Rellin, L.; Heeren, J.; Beisiegel, U. Recycling of Apolipoprotein E Is Not Associated with Cholesterol Efflux in Neuronal Cells. Biochim. Biophys. Acta—Mol. Cell Biol. Lipids 2008, 1781, 232–238.
  91. Lindner, K.; Beckenbauer, K.; van Ek, L.C.; Titeca, K.; de Leeuw, S.M.; Awwad, K.; Hanke, F.; Korepanova, A.V.; Rybin, V.; van der Kam, E.L.; et al. Isoform- and Cell-State-Specific Lipidation of ApoE in Astrocytes. Cell Rep. 2022, 38, 110435.
  92. Minagawa, H.; Gong, J.S.; Jung, C.G.; Watanabe, A.; Lund-Katz, S.; Phillips, M.C.; Saito, H.; Michikawa, M. Mechanism Underlying Apolipoprotein E (ApoE) Isoform-Dependent Lipid Efflux From Neural Cells in Culture. J. Neurosci. Res. 2009, 87, 2498.
  93. Rawat, V.; Wang, S.; Sima, J.; Bar, R.; Liraz, O.; Gundimeda, U.; Parekh, T.; Chan, J.; Johansson, J.O.; Tang, C.; et al. ApoE4 Alters ABCA1 Membrane Trafficking in Astrocytes. J. Neurosci. 2019, 39, 9611–9622.
  94. Chan, H.C.; Ke, L.Y.; Lu, H.T.; Weng, S.F.; Chan, H.C.; Law, S.H.; Lin, I.L.; Chang, C.F.; Lu, Y.H.; Chen, C.H.; et al. An Increased Plasma Level of ApoCIII-Rich Electronegative High-Density Lipoprotein May Contribute to Cognitive Impairment in Alzheimer’s Disease. Biomedicines 2020, 8, 542.
  95. Yassine, H.N.; Feng, Q.; Chiang, J.; Petrosspour, L.M.; Fonteh, A.N.; Chui, H.C.; Harrington, M.G. ABCA1-Mediated Cholesterol Efflux Capacity to Cerebrospinal Fluid Is Reduced in Patients With Mild Cognitive Impairment and Alzheimer’s Disease. J. Am. Heart Assoc. 2016, 5, e002886.
  96. Marchi, C.; Adorni, M.P.; Caffarra, P.; Ronda, N.; Spallazzi, M.; Barocco, F.; Galimberti, D.; Bernini, F.; Zimetti, F. ABCA1-And ABCG1-Mediated Cholesterol Efflux Capacity of Cerebrospinal Fluid Is Impaired in Alzheimer’s Disease. J. Lipid Res. 2019, 60, 1449–1456.
  97. Cipollari, E.; Szapary, H.J.; Picataggi, A.; Billheimer, J.T.; Lyssenko, C.A.; Ying, G.S.; Shaw, L.M.; Kling, M.A.; Kaddurah-Daouk, R.; Rader, D.J.; et al. Correlates and Predictors of Cerebrospinal Fluid Cholesterol Efflux Capacity from Neural Cells, a Family of Biomarkers for Cholesterol Epidemiology in Alzheimer’s Disease. J. Alzheimer’s Dis. 2020, 74, 563–578.
  98. Li, X.; Zhang, J.; Li, D.; He, C.; He, K.; Xue, T.; Wan, L.; Zhang, C.; Liu, Q. Astrocytic ApoE Reprograms Neuronal Cholesterol Metabolism and Histone-Acetylation-Mediated Memory. Neuron 2021, 109, 957–970.
  99. Zimetti, F.; Caffarra, P.; Ronda, N.; Favari, E.; Adorni, M.P.; Zanotti, I.; Bernini, F.; Barocco, F.; Spallazzi, M.; Galimberti, D.; et al. Increased PCSK9 Cerebrospinal Fluid Concentrations in Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 55, 315–320.
  100. Picard, C.; Poirier, A.; Bélanger, S.; Labonté, A.; Auld, D.; Poirier, J. Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) in Alzheimer’s Disease: A Genetic and Proteomic Multi-Cohort Study. PLoS ONE 2019, 14, e0220254.
  101. Rauch, J.N.; Luna, G.; Guzman, E.; Audouard, M.; Challis, C.; Sibih, Y.E.; Leshuk, C.; Hernandez, I.; Wegmann, S.; Hyman, B.T.; et al. LRP1 Is a Master Regulator of Tau Uptake and Spread. Nature 2020, 580, 381.
More
Video Production Service