Abstract

Supramolecular protein assemblies have been used as intelligent drug delivery systems that can encapsulate drugs and transport them to specific tissues or cells. However, the known methods for designing supramolecular protein assemblies for transportation across the blood-brain barrier (BBB) remain challenging and inefficient. Herein, we report that the supramolecular recombinant-protein-based strategy enables the biosynthesis and production of a supramolecular protein assembly that is intrinsically capable of crossing the BBB. The recombinant protein constituting the essential part of apolipoprotein A1 can self-assemble into a supramolecular protein assembly known as a nanodisc. The nanodisc could efficiently enter the brain of an Alzheimer’s disease mouse model, recognize Aβ1–42, eliminate amyloid plaques, promote neurogenesis, and ameliorate cognitive impairment. This work opens a new field for supramolecular protein assemblies and offers a new avenue for designing versatile and intelligent supramolecular biomaterials.
Keywords: Alzheimer’s disease, blood-brain barrier, nanodisc, nanomedicine, protein assembly
Alzheimer’s disease (AD) is a neurodegenerative disorder that primarily affects memory and cognitive function.1,2 It is estimated that more than 50 million individuals worldwide have AD or dementia. This is expected to triple by 2050, owing to global population aging.3 The limited understanding of its underlying mechanisms poses a great challenge in its management.4 Although significant progress has been achieved in understanding the biological mechanisms underlying AD pathogenesis, its exact cause and progression are not yet fully understood.5 Therefore, developing effective and targeted therapeutic agents remains a challenge.6 Moreover, the failure rate of clinical trials for AD is approximately 100%, owing to the complex pathological mechanisms and low penetration of drugs across the blood-brain barrier (BBB).7 Therefore, novel therapeutic strategies are urgently needed.8−13
Various supramolecular entities have been studied for their potential applications in AD treatment.14−18 In addition to cyclodextrins,19 other classes of macrocyclic compounds, including calixarenes,20 cucurbit[n]urils,21,22 crown ethers,23,24 and other molecular containers,25,26 have been investigated. Although these supramolecular macrocycles play a vital role in self-assembly, several factors restrict their use in treating AD. First, these macrocyclic compounds often cannot cross the BBB independently.27,28 Second, some of these compounds lack target specificity or multitarget capability for neurons.29−31 Lastly, macrocycles themselves do not generally possess a therapeutic function,32,33 and biocompatibility remains a major concern.34
Protein assemblies with BBB-crossing capability may provide an alternative platform for designing novel drug carrier systems into the central nervous system (CNS).35−37 Protein-based biomaterials are biocompatible and functionally diverse.38,39 Furthermore, recombinant protein technology could generate proteins in substantial quantities by utilizing genetically modified organisms, including bacteria or yeast.40−45 Many proteins, such as lipoproteins, are closely associated with AD pathology;46,47 they can be transported across the BBB through diverse receptor-mediated transcytosis mechanisms.48−50 Therefore, engineering protein-based supramolecular assemblies carrying molecules essential for lipoproteins to cross the BBB may provide an ideal platform for curing AD.
Nanodiscs are supramolecular protein assemblies comprising phospholipids and encircled amphipathic proteins with helical belts (Figure 1).51 Nanodiscs are widely used to study membrane proteins since they can solubilize and stabilize membrane proteins and replicate a more natural environment than liposomes; this is because their phospholipid bilayer with a hydrophobic edge is screened utilizing two amphipathic proteins.52,53 The structure of nanodiscs is comparable to that of the discoidal high-density lipoprotein (HDL). Apolipoprotein A1 (ApoA1), a major protein component of HDLs, was modified to form an ApoA1-based supramolecular recombinant protein that stabilizes nanodiscs.54,55 Recently, a study revealed that ApoA1 can lower the risk of AD and provide protection by exhibiting amyloid beta (Aβ) affinity.56 Therefore, nanodiscs could be used as a neuroprotective supramolecular protein assembly in AD therapy.57
Figure 1.
Supramolecular recombinant protein (SRP)-based strategy enables the biosynthesis and production of a protein assembly that can intrinsically cross the BBB and capture Aβ, facilitating its degradation by microglial cells, thereby inhibiting the aberrant production of Aβ. This schematic diagram shows the structural considerations needed for the biosynthesis of the nanodisc and its BBB-crossing and AD-related abilities.
In this study, nanodiscs [comprising 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) and SRP-A] exhibited efficient BBB-crossing ability and promoted the uptake of Aβ by microglia for clearance, which are important requirements for treating AD. A general strategy for engineering a supramolecular protein assembly with intrinsic BBB-crossing ability was also devised, which is named the supramolecular recombinant protein (SRP)-based strategy. Based on a recombinant protein design approach, the SRP strategy endows protein-based macrocycles with intrinsic BBB-crossing ability and facilitates AD-related functions. We explored the possibility of nanodiscs as therapeutic reagents in vitro and in vivo, evaluated their targeted ability toward Aβ, how they inhibit the fibrillation and detaching of fibrils, reduce oligomer populations and cytotoxicity against Aβ fibrils, enhance microglial phagocytosis of Aβ, and rectify cognitive dysfunction in an AD mouse model (Figure 1). These findings will help to design novel supramolecular protein assemblies and therapeutic agents for AD.
ApoA1 has been suggested to be involved in certain neurological functions and disorders. Although the detailed mechanism underlying the movement of ApoA1 across the BBB remains elusive, some studies indicated that this movement involves the binding of ApoA1 to specific transporters, such as scavenger receptor class B type I (SR-BI), which is expressed in the brain and the liver.56 To explore the ability of SRP-fabricated macrocycles to cross the BBB, we used a nanodisc with an ApoA1 component as an SRP-A. The SRP-A retained the key tandem amphipathic α-helices 1–10 (designated as H1–H10; see Supporting Information Figure S1a) in the human ApoA1 structure, which are responsible for binding to the lipid bilayer. DYDIPTT (marked in orange in Figure S1a) is the linker between the hepta-His-tag and the TEV protease recognition site, ENLYFQG. This linker stabilizes the structure of the nanodisc, owing to its amphipathic helical properties.58,59 The His-tagged section allows large-scale SRP-A production.60,61 For the recombinant production of ApoA1, SRP-A inherits the interhelical salt bridge of ApoA1 so that the two SRP-As interact in an antiparallel conformation (Supporting Information Figure S1b). SRP-A was expressed in Escherichia coli and induced by isopropyl β-d-1-thiogalactopyranoside; thereafter, it was purified through a Ni-NTA affinity column. SDS-PAGE revealed a single protein band at ∼26 kDa, suggesting the successful purification of SRP-A (Supporting Information Figure S2b). After mixing SRP-A with lipids in a specific ratio and purifying using BioBeads, the self-assembled nanodiscs formed the transparent solution (Figure 2a). Size exclusion chromatography revealed only one peak corresponding to the purified product (Figure 2b). Dynamic light scattering (DLS) analysis revealed that the size of the nanodiscs was 10 nm (Figure 2c). Zeta potential measurements revealed that the nanodiscs were relatively neutral (ζ = −0.2 mV). Transmission electron microscopy (TEM) revealed that the diameter of the nanodiscs was ∼10 nm (Figure 2d,e), and atomic force microscopy revealed that the height of the nanodiscs was approximately 7 nm (Figure 2f), similar to the height of lipid bilayers. Collectively, these results suggest the successful establishment of the desired nanodiscs. The precise modification of certain atoms is also possible for this protein assembly. Here, the site mutation of the nanodisc at position 56 from Ser to Cys [SRP-A (S56C)] allowed further dye labeling for easy visualization. This mutation had a negligible effect on the nanodisc structure (Supporting Information Figure S3).
Figure 2.
(a) Macroscopic photograph of the mixture of SRP-A and DMPC before assembly and the assembled nanodiscs in PBS. (b) SEC characterization of the nanodiscs monitored at λ = 280 nm. (c) Particle size distribution of the nanodiscs analyzed via DLS analysis. (d) Morphology of the nanodiscs, as observed using TEM. (e) Higher magnification of the TEM image in (d). (f) AFM morphology and height profile of the nanodiscs. (g) Schematic of the experimental design for blood-brain barrier (BBB) permeability study of the nanodiscs. Brain distribution of the (h) IAF-nanodiscs and (i) BODIPY-nanodisc after intravenous administration (n = 3). (j) Selenium (Se) content in the saline and selenocystine-mutated nanodisc (S56U), respectively (n = 3). (k) Selenium content in the brain tissues of mice treated with the saline and nanodisc (S56U) (n = 3). (l) Images showing the distribution of BODIPY-nanodiscs in the cortex and hippocampus of AD mice (APP/PS1) taken using confocal microscopy of frozen brain sections at 4 h after the intravenous administration of BODIPY-nanodiscs. Nuclei are stained blue (DAPI: 4′,6-diamidino-2-phenylindole), BODIPY-nanodiscs are stained red, and Aβ aggregates are stained green (FAM: fluorescein amidite) (n = 7).
Using two distinct labeling strategies, we also investigated whether the nanodiscs could cross the BBB after systemic intravenous delivery. First, using SRP-A (S56C), we labeled the nanodisc with the fluorescent dye 5-iodoacetamidofluorescein (5-IAF) via covalent conjugation of the thiol-iodoacetamide groups. Fluorescein is commonly used for labeling and detection in various biological applications. A 20 μM solution (200 μL) of 5-IAF-labeled nanodiscs (referred to as 5-IAF-nanodiscs) was administered to BALB/c mice via the tail vein. After 4 h, the mice were euthanized and perfused with saline. Then, the skull was removed, and the organs were visualized by using an IVIS imaging system at an excitation wavelength of 493 nm. Figure 2h (bottom row) illustrates that the brain of mice treated with 5-IAF-nanodiscs exhibited strong fluorescence at 4 h postinjection; different from the brains of mice treated with 5-IAF and saline alone, in which no fluorescent signal was detected (middle row in Figure 2h). Interestingly, when utilizing 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindotricarbocyanine iodide (BODIPY;62 excitation wavelength, 502 nm) as the covalently conjugated dye under similar conditions, the brains of mice treated with BODIPY-nanodiscs exhibited a stronger BODIPY fluorescence signal compared with those treated with the BODIPY dye or saline alone, wherein no fluorescent signal was observed (Figure 2i). This result further confirms that nanodiscs can traverse the BBB irrespective of the type of conjugated dye used.
To evaluate the efficiency of nanodiscs in crossing the BBB, we developed a selenium-mutated nanodisc (S56U) using a cysteine auxotrophic expression system, in which cysteine (Cys) was biosynthetically substituted with selenocysteine (Sec, U) (Supporting Information Figure S5). This allows us to track crossing efficiency by monitoring selenium levels. The BBB permeability of the selenocysteine-containing nanodiscs was assessed using inductively coupled plasma mass spectrometry (ICP-MS), with the expectation that selenium levels in the brain would exceed those in normal tissue if the nanodiscs successfully cross the BBB. As shown in Figure 2j, the nanodisc (S56U) represents the selenium-containing nanodisc derived from site-mutated SRP-A (S56C) expressed through the cysteine auxotrophic system. In in vitro testing, the Se content of Nanodisc (S56U) was measured at 199.13 μg/L, compared to 5.45 μg/L in the control saline, resulting in an increase of 193.68 μg/L. Figure 2k shows that after tail vein injection of Nanodisc (S56U) in mice, the Se concentration in brain samples increased from 7.90 to 30.76 μg/L, an increase of 22.86 μg/L. This indicates an approximate BBB-crossing efficiency of 11.8%. These findings conclusively demonstrate the nanodisc’s significant intrinsic ability to cross the BBB efficiently.
We also observed that the BODIPY-nanodiscs were distributed in the cortex and hippocampus of the nanodisc-injected AD mouse model (APP/PS1). Figure 2l shows mouse cortex and hippocampus immunostaining images after nanodisc treatment. The red BODIPY-nanodiscs were distributed extensively around the Aβ deposits, which were stained green. After the in vivo evaluation of BODIPY-nanodisc localization over 4 h, in vitro studies were undertaken to determine whether the observed uptake was due to accumulation or binding. As shown in the TEM images in Supporting Information Figure S6, Aβ1–42 oligomers bound to the lateral side of the nanodiscs, consistent with the confocal laser scanning microscopy (CLSM) results. The binding affinity (Kd) for Aβ1–42 with nanodiscs was determined to be 1.36 μM (Supporting Information Figure S7). To assess the efficiency of the assembled nanodisc versus individual proteins (SRP-A) during crossing the BBB, we measured the fluorescence intensity in brain tissue samples at 1 and 4 h postinjection in mice (Supporting Information Figure S8a). The results showed that the assembled nanodisc exhibited 48% higher fluorescence intensity than SRP-A at 1 h and 58% higher fluorescence intensity at 4 h postinjection (Supporting Information Figure S8b), indicating significantly improved BBB penetration for the protein assemblies. Furthermore, to verify that nanodiscs target monocytes via the SR-B1 pathway, we performed inhibition experiments using HDL to block nanodisc uptake. A competitive inhibition experiment further confirmed that nanodiscs cross the BBB via an SR-BI receptor-mediated transcytosis process (Supporting Information Figure S9). High concentrations of HDL reduced nanodisc (5-IAF) uptake by 39.1% under 20-fold excess HDL, validating the SR-B1-targeting capability of nanodiscs for specific delivery to monocytes in peripheral blood. These results further confirmed that nanodiscs can effectively cross the BBB and play a potential therapeutic role in brain-targeted AD therapy.
Uptake and degradation by microglia are the key mechanisms underlying Aβ clearance in the brain. Microglia can clear Aβ via uptake or phagocytosis and capture large Aβ deposits in plaques, minimizing damage to adjacent nerves.63 Therefore, to assess the effect of nanodiscs on internal cerebral Aβ degradation, microglia were cultured and incubated with soluble Aβ1–42 and nanodiscs to evaluate the cellular uptake, distribution, and degradation of Aβ1–42. Using CLSM, we studied the uptake of FAM-labeled Aβ1–42 by BV-2 cells with nanodiscs. The colocalization of Aβ with lysosomes in the presence of nanodiscs after a short incubation time (15 min) indicated the lysosomal transport of Aβ (Figure 3a,c). As shown in Figure 3d, ELISA revealed that the cells treated with nanodiscs had intracellular Aβ concentrations significantly lower than those of the control group, which was treated with Aβ only. In particular, the intracellular Aβ concentration was 149.4 ± 3.067 pg/mg protein in nanodisc-treated cells; it was 391.1 ± 1.087 pg/mg protein in the control group. These findings indicate that Aβ degradation by microglial cells is enhanced in the presence of the nanodiscs.
Figure 3.
Effect of nanodiscs on the uptake, cellular distribution, and degradation of amyloid beta (Aβ)1–42 by microglia and Chang liver cells. After 15 min of incubation, the cellular absorption and distribution of FAM-labeled Aβ1–42 (FAM-Aβ) in (a) microglia or (b) Chang liver cells are shown in the presence or absence of nanodiscs, n = 7. Lysosomes were stained with LysoTracker Red. The red arrows indicated that FAM-Aβ colocalized with lysosomes. The white arrow indicated that FAM-Aβ did not colocalize with lysosomes. Scale bar: 10 μm. (c, e) Co-localization coefficient of FAM-Aβ to lysosomes. (d, f) After 3 h of incubation with Aβ or Aβ + nanodiscs, the intracellular Aβ1–42 concentration was measured and normalized to the total protein concentration (the final concentration of the nanodiscs is 1.8 μM). Data are presented as mean ± SD and were tested by using an ordinary one-way t test. Results are representative of three biological replicates. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 compared with treatment with Aβ only.
The liver plays a key role in the degradation and clearance of Aβ from the peripheral blood, which in turn regulates how much Aβ crosses into the brain.63,64 Additionally, the liver produces apolipoproteins, such as ApoA1, which influence Aβ aggregation and clearance. Evaluating liver cells helps us understand these clearance mechanisms, including interactions with transporters like SR-BI, which are expressed in both the liver and brain.56 Therefore, Chang liver cells were used in this study to mimic hepatocytes and determine the effect of nanodiscs on the Aβ cellular uptake and clearance in the liver. Similar to microglia, the presence of nanodiscs enhanced the cellular absorption of Aβ (Figure 3b). Moreover, more Aβ appeared to be colocalized with lysosomes (Figure 3e). After 3 h of incubation, more Aβ was degraded in the group treated with nanodiscs. ELISA detection revealed intracellular Aβ concentrations of 0.522 ± 0.325 and 2.822 ± 0.493 pg/mg protein in the groups treated with or without nanodiscs, respectively (Figure 3e). This could be explained by the strong Aβ1–42-binding affinity of the nanodiscs. Collectively, these findings suggest that the nanodiscs enhanced the cellular absorption, intracellular localization, and degradation of Aβ1–42 monomers/oligomers in the microglia, supporting the idea that the ApoA1-originated SRP possessed a significant Aβ clearance ability.
The spatial learning and memory of mice treated with or without nanodiscs were evaluated by using the Morris water maze (MWM) test (Figure 4). Transgenic APP/PS1 mice exhibiting selective enhancement of Aβ1–42 and plaque deposition (Figure 4a) are suitable for this study65 and were selected for behavioral assessment. On the first day, nanodisc-injected APP/PS1 mice (120 s) displayed no significant difference in escape latency (Figure 4b) compared to normal and saline-injected APP/PS1 mice. On the second day, the escape latency of nanodisc-injected APP/PS1 mice was 73 s, whereas it was 120 s for AD mice treated with saline (Figure 4b). A similar trend was observed on days 3 and 4, demonstrating a significant improvement in the performance of the mice after administration. On the last day of training, a memory study was conducted with the platform removed. The time spent in the targeted quadrant (%) was higher for nanodisc-injected APP/PS1 mice (27%) than for saline-treated APP/PS1 mice (17%) (Figure 4c). Furthermore, we observed that the nanodisc-injected mice presented remarkable progress in searching strategies to find the platform (Figure 4d,e). Moreover, the number of nanodisc-treated APP/PS1 mice crossing the target site was three times more, showing a higher percentage of platform crossings in the target quadrant than saline-treated APP/PS1 (0 times) or normal mice (0.5 times, the average number of times for all mice in the group). These results imply that the nanodiscs exhibited potential therapeutic effects on APP/PS1 mice, which conserved their spatial memory. All the above experimental results indicate that after 4 weeks of consecutive tail vein injection of nanodiscs, the spatial learning and memory ability of AD model mice improved, indicating the memory-rescue effect of the nanodiscs.
Figure 4.

Nanodiscs rescued memory deficits in APP/PS1 mice (n = 8). (a) Treatment schedule, pathological monitoring, and therapeutic evaluation. (b) Escape latency at different time points. (c) Percentage of time spent in the quadrant where the escape platform was located. (d) Number of times of platform crossing on the final day with the platform removed. (e) Representative swimming paths of the mice in the Morris water maze after different treatments. Data are presented as mean ± SD and were analyzed using the t test. *p < 0.05, **p < 0.01, ***p < 0.001 compared with APP/PS1 mice treated with normal saline.
The effect of nanodiscs in reducing Aβ deposition was assessed by using immunohistochemistry. A drastic difference in Aβ deposition was observed between APP/PS1 and C57BL/6J mice (Figure 5a,c). Anti-Aβ immunostaining revealed that, compared with saline-injected APP/PS1 mice, the amount of amyloid plaques was considerably reduced in the cortex and hippocampus of mice treated with 20 μM nanodiscs (200 μL of tail vein injection per day for 2 weeks). Anti-Aβ immunostaining also revealed that compared with the saline-injected APP/PS1 mice, amyloid plaque loads were reduced by 47% and 63% in the cortex and by 52% and 57% in the hippocampus after injection with 20 μM nanodiscs, respectively (Figure 5c). These results further confirm that the nanodiscs exhibited a therapeutic effect on the APP/PS1 mice. In particular, the inhibitory effect of the nanodiscs on amyloid plaques was significantly evident at a concentration of 20 μM. Moreover, Aβ deposition was notably decreased in nanodisc-injected APP/PS1 mice, confirming the Aβ clearance capacity of the nanodiscs.
Figure 5.

Nanodiscs (a, c) reduced Aβ deposition and (b, d) attenuated microgliosis. (e) Nissl staining of the neurons in the cortex and hippocampus of C57BL/6J mice or AD mice treated daily with saline or nanodiscs for 4 weeks. Immunostaining was performed on 4 mm thick brain sections using anti-Aβ or anti-CD45 antibodies. Data are presented as the mean ± SD. Data were analyzed using one-way analysis of variance (Dunnett’s multiple comparisons test) by comparing each treatment group with the APP/PS1 + saline group. n = 8. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 compared with saline-treated APP/PS1 mice.
Abnormal microglial activation has been discovered in the brains of patients with AD and mouse models of amyloidosis.66 Previous research suggested that Aβ oligomers and fibrils activate neuroinflammatory pathways.67 Herein, we assessed microglial activation using CD45 as a marker. Compared with saline-injected APP/PS1 mice, approximately 62% and 51% lower CD45-positive activated microglial load was observed in the cortex and hippocampus, respectively, of mice treated with 20 μM nanodiscs (Figure 5b,d). Since Aβ aggregates play a crucial role in microglial activation in AD brains, the decreased number of activated microglia observed in mice treated with nanodisc injections may be due to the ability of the nanodiscs to remove Aβ.
A defining feature of AD is neuronal loss in the cortex and hippocampus.68 The cytoplasm can be stained with toluidine blue as Nissl granules in neuronal cell bodies. Comparing saline-injected animals with APP/PS1 mice, Nissl staining revealed that the cortex and hippocampus displayed neuronal hypocellularity and nuclear shrinkage. Nanodisc administration for 2 weeks considerably attenuated the damage to neuronal integrity and neuronal loss in APP/PS1 mice (Figure 5e). Furthermore, APP/PS1 mice injected with 20 μM nanodiscs for 2 weeks exhibited significantly better improvements in cortical cell atrophy than the untreated group and significantly more cells in the hippocampal region than the disease model group. Cell atrophy disappeared in the cerebral cortex of APP/PS1 mice injected intravenously with 20 μM nanodiscs for 2 weeks. Moreover, the number of cells in the hippocampal region was significantly higher than that in the disease model group and almost equal to that in normal mice. The results of Nissl staining suggest that APP/PS1 mice administered with nanodiscs for 2 weeks exhibited significantly improved cellular atrophy in the cerebral cortex than the untreated group; this trend was more obvious upon injection with 20 μM nanodiscs. Hence, nanodisc treatment significantly reduced the damage to neuronal integrity and neuronal loss in APP/PS1 mice. This may be because nanodiscs regulate antioxidant stress through the Nrf2 signaling pathway, inducing an antioxidant response in vivo. This mechanism facilitates the clearance of Aβ and mitigates neuroinflammation within the brain, while exhibiting no impact on the regular lipid metabolism process (Supporting Information Figure S10).
To assess the biosafety of the nanodiscs in treating AD, the primary organs were stained with hematoxylin and eosin (H&E). After 4 weeks of nanodisc injection, histological sections of the heart, liver, spleen, lungs, and kidneys exhibited no obvious organ damage or inflammation (Supporting Information Figure S11). Inflammatory responses such as alveolar septal hemorrhage, peri-alveolar capillary congestion, and interstitial focal lymphocyte infiltration were observed in the lungs of APP/PS1 mice that received saline injections, as previously reported. No such pathological changes appeared in the nanodisc-injected group. Therefore, the in vivo injection of nanodiscs is generally safe at the dose administered in the present study. Additionally, flow cytometry was performed on C57BL/6 mice injected with nanodiscs to assess immunogenicity. After 4 h, CD3+CD4+ T-cell activation was analyzed in the blood, spleen, and lymph nodes (Supporting Information Figure S12). The results indicated a modest increase in CD3+CD4+ T cells in the blood (*p < 0.05), suggesting a mild immune response. No significant differences were observed in the spleen or lymph nodes (*p > 0.05). This modest increase in blood T cells is consistent with normal immune responses observed in similar studies,69 supporting the low systemic immunogenicity of nanodiscs and their suitability for potential therapeutic use.
In conclusion, the distinct and increased BBB-crossing ability and therapeutic performance of the nanodiscs against AD in a biological environment have generated new interest in the classic supramolecular protein assembly. Our in vitro and in vivo experiments revealed promising disease-ameliorating effects. Nanodiscs accelerated microglial-cell-induced Aβ degradation by promoting their lysosomal transport. After intravenous injection, the nanodiscs decreased amyloid deposits, attenuated microgliosis, improved neurological damage, and rescued memory decline in AD mouse models. The key principle of the SRP strategy in bestowing a supramolecular protein assembly with an intrinsic ability to cross the BBB is vastly underestimated for classical organic self-assemblies. This strategy provides a potential therapeutic effect for nanodiscs against AD, which is attributable to the ApoA1 scaffold. Accordingly, it could be proposed that with the rapidly growing knowledge about receptor-mediated transcytosis, more BBB-crossing proteins (e.g., transferrins, insulins, and other lipoproteins) can be utilized in designing supramolecular assemblies via SRP and can be used as potential brain delivery and/or therapeutic agents for CNS diseases.
Acknowledgments
We thank the Analytical & Testing Center of NPU for material characterization. All animal experiments were conducted according to the National Institute of Health Guide for the Care and Use of Laboratory Animals and approved by the Northwestern Polytechnical University Ethics Committee (No. 202001027). Y. G. is an inventor on a patent application related to this work filed by Northwestern Polytechnical University (no. 202210510621.9, filed 20 May 2022).
Glossary
Abbreviations
- AD
Alzheimer’s disease
- BBB
blood-brain barrier
- CLSM
confocal laser scanning microscopy
- CNS
central nervous system
- DLS
dynamic light scattering
- H&E
hematoxylin and eosin
- HDL
high-density lipoprotein
- MWM
Morris water maze
- SRP
supramolecular recombinant protein
- TEM
transmission electron microscopy
- WT
wild-type
Supporting Information Available
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.nanolett.4c03672.
Materials and methods, sequence analysis, purification, domains interaction analysis, TEM, effect of Lipid-to-SRP-A ratio, identification of selenium in nanodiscs, SPR measurements, BBB crossing efficiency, competitive inhibitin experiments, antioxidant stress and response, biosafety, additional references (PDF)
Author Present Address
∥ Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, 518055, China
Author Contributions
+ Y.H. and X.S. contributed equally to this work. Y.G. and Z.Q. conceived the concept for this work. Y.H. and X.S. contributed to the preparation and measurement of protein samples, as well as analysis of the data. Z.C. and X.W. assisted with the preparation of protein nanodiscs. Z.Y. assisted with the characterization. F.C. and B.D. provided assistance. J.L. and X.D. assisted with the in vivo bioevaluation of nanodiscs. Y.G., Y.H., X.S., R.W., Z.Q., and Y.G. collectively wrote the manuscript.
We gratefully acknowledge financial support from the National Natural Science Foundation of China (22007078, 22071196, 22471220), Key R&D Program of Shaanxi Province (2021KWZ-18), Student Innovation and Entrepreneurship Education Center of the Student Work Department of the Party Committee of NPU (2021-cxcy-012), Higher Education Research Fund of NPU (CJGZMS202202), the Fundamental Research Funds for the Central Universities, and Fellowship from CSC Innovative Team Program (CXXM2110141862).
The authors declare no competing financial interest.
Supplementary Material
References
- Knopman D. S.; Amieva H.; Petersen R. C.; Chetelat G.; Holtzman D. M.; Hyman B. T.; Nixon R. A.; Jones D. T. Alzheimer disease. Nat. Rev. Dis. Primers 2021, 7, 33. 10.1038/s41572-021-00269-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leng F.; Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here?. Nat. Rev. Neurol. 2021, 17, 157. 10.1038/s41582-020-00435-y. [DOI] [PubMed] [Google Scholar]
- Yang A. C.; Stevens M. Y.; Chen M. B.; Lee D. P.; Stahli D.; Gate D.; Contrepois K.; Chen W.; Iram T.; Zhang L.; Vest R. T.; Chaney A.; Lehallier B.; Olsson N.; du Bois H.; Hsieh R.; Cropper H. C.; Berdnik D.; Li L.; Wang E. Y.; Traber G. M.; Bertozzi C. R.; Luo J.; Snyder M. P.; Elias J. E.; Quake S. R.; James M. L.; Wyss-Coray T. Physiological blood-brain transport is impaired with age by a shift in transcytosis. Nature 2020, 583, 425. 10.1038/s41586-020-2453-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- El-Hayek Y. H.; Wiley R. E.; Khoury C. P.; Daya R. P.; Ballard C.; Evans A. R.; Karran M.; Molinuevo J. L.; Norton M.; Atri A. Tip of the Iceberg: Assessing the Global Socioeconomic Costs of Alzheimer’s Disease and Related Dementias and Strategic Implications for Stakeholders. JAD 2019, 70, 323. 10.3233/JAD-190426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Graff-Radford J.; Yong K. X. X.; Apostolova L. G.; Bouwman F. H.; Carrillo M.; Dickerson B. C.; Rabinovici G. D.; Schott J. M.; Jones D. T.; Murray M. E. New insights into atypical Alzheimer’s disease in the era of biomarkers. Lancet Neurol. 2021, 20, 222. 10.1016/S1474-4422(20)30440-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Congdon E. E.; Sigurdsson E. M. Tau-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 399. 10.1038/s41582-018-0013-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu P. P.; Xie Y.; Meng X. Y.; Kang J. S. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduct. Target Ther. 2019, 4, 29. 10.1038/s41392-019-0063-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Q.; Song Q.; Gu X.; Zheng M.; Wang A.; Jiang G.; Huang M.; Chen H.; Qiu Y.; Bo B.; Tong S.; Shao R.; Li B.; Wang G.; Wang H.; Hu Y.; Chen H.; Gao X. Multifunctional Nanostructure RAP-RL Rescues Alzheimer’s Cognitive Deficits through Remodeling the Neurovascular Unit. Adv. Sci. 2021, 8, 2001918 10.1002/advs.202001918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai L.; Yang C.; Jia W.; Liu Y.; Xie R.; Lei T.; Yang Z.; He X.; Tong R.; Gao H. Endo/lysosome-escapable delivery depot for improving BBB transcytosis and neuron targeted therapy of alzheimer’s disease. Adv. Funct. Mater. 2020, 30, 1909999 10.1002/adfm.201909999. [DOI] [Google Scholar]
- a Han G.; Bai K.; Yang X.; Sun C.; Ji Y.; Zhou J.; Zhang H.; Ding Y. ″Drug-Carrier″ Synergy Therapy for Amyloid-β Clearance and Inhibition of Tau Phosphorylation via Biomimetic Lipid Nanocomposite Assembly. Adv. Sci. 2022, 9, e2106072 10.1002/advs.202106072. [DOI] [PMC free article] [PubMed] [Google Scholar]; b Zhao Y.; Cai J.; Liu Z.; Li Y.; Zheng C.; Zheng Y.; Chen Q.; Chen H.; Ma F.; An Y.; Xiao L.; Jiang C.; Shi L.; Kang C.; Liu Y. Nanocomposites Inhibit the Formation, Mitigate the Neurotoxicity, and Facilitate the Removal of β-Amyloid Aggregates in Alzheimer’s Disease Mice. Nano Lett. 2019, 19, 674–683. 10.1021/acs.nanolett.8b03644. [DOI] [PubMed] [Google Scholar]; c Jung M.; Kim H.; Hwang J. W.; Choi Y.; Kang M.; Kim C.; Hong J.; Lee N. K.; Moon S.; Chang J. W.; Choi S. J.; Oh S. Y.; Jang H.; Na D. L.; Kim B. S. Iron Oxide Nanoparticle-Incorporated Mesenchymal Stem Cells for Alzheimer’s Disease Treatment. Nano Lett. 2023, 23, 476–490. 10.1021/acs.nanolett.2c03682. [DOI] [PubMed] [Google Scholar]; d Tracy G. C.; Huang K. Y.; Hong Y. T.; Ding S.; Noblet H. A.; Lim K. H.; Kim E. C.; Chung H. J.; Kong H. Intracerebral Nanoparticle Transport Facilitated by Alzheimer Pathology and Age. Nano Lett. 2023, 23, 10971–10982. 10.1021/acs.nanolett.3c03222. [DOI] [PMC free article] [PubMed] [Google Scholar]; e Anraku Y.; Kuwahara H.; Fukusato Y.; Mizoguchi A.; Ishii T.; Nitta K.; Matsumoto Y.; Toh K.; Miyata K.; Uchida S.; Nishina K.; Osada K.; Itaka K.; Nishiyama N.; Mizusawa H.; Yamasoba T.; Yokota T.; Kataoka K. Glycaemic control boosts glucosylated nanocarrier crossing the BBB into the brain. Nat. Commun. 2017, 8, 1001. 10.1038/s41467-017-00952-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang X.; Yang W.; Xia X.; Lei T.; Yang Z.; Jia W.; Zhou Y.; Cheng G.; Gao H. Intranasal Delivery of BACE1 siRNA and Rapamycin by Dual Targets Modified Nanoparticles for Alzheimer’s Disease Therapy. Small 2022, 18, e2203182 10.1002/smll.202203182. [DOI] [PubMed] [Google Scholar]
- Ma M.; Gao N.; Li X.; Liu Z.; Pi Z.; Du X.; Ren J.; Qu X. A Biocompatible Second Near-Infrared Nanozyme for Spatiotemporal and Non-Invasive Attenuation of Amyloid Deposition through Scalp and Skull. ACS Nano 2020, 14, 9894. 10.1021/acsnano.0c02733. [DOI] [PubMed] [Google Scholar]
- Xu Z.; Jia S.; Wang W.; Yuan Z.; Jan Ravoo B.; Guo D. S. Heteromultivalent peptide recognition by co-assembly of cyclodextrin and calixarene amphiphiles enables inhibition of amyloid fibrillation. Nat. Chem. 2019, 11, 86. 10.1038/s41557-018-0164-y. [DOI] [PubMed] [Google Scholar]
- Wang J.; Shangguan P.; Chen X.; Zhong Y.; Lin M.; He M.; Liu Y.; Zhou Y.; Pang X.; Han L.; Lu M.; Wang X.; Liu Y.; Yang H.; Chen J.; Song C.; Zhang J.; Wang X.; Shi B.; Tang B. Z. A one-two punch targeting reactive oxygen species and fibril for rescuing Alzheimer’s disease. Nat. Commun. 2024, 15, 705. 10.1038/s41467-024-44737-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G.; Li Y. M. Modulating the aggregation of amyloid proteins by macrocycles. Aggregate 2022, 3, e161 10.1002/agt2.161. [DOI] [Google Scholar]
- Consoli G. M. L.; Tosto R.; Baglieri A.; Petralia S.; Campagna T.; Di Natale G.; Zimbone S.; Giuffrida M. L.; Pappalardo G. Novel Peptide-Calix[4]arene Conjugate Inhibits Aβ Aggregation and Rescues Neurons from Aβ’s Oligomers Cytotoxicity In Vitro. ACS Chem. Neurosci. 2021, 12, 1449. 10.1021/acschemneuro.1c00117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan Y. C.; Hu X. Y.; Guo D. S. Biomedical Applications of Calixarenes: State of the Art and Perspectives. Angew. Chem., Int. Ed. 2021, 60, 2768. 10.1002/anie.201916380. [DOI] [PubMed] [Google Scholar]
- Furtado D.; Bjornmalm M.; Ayton S.; Bush A. I.; Kempe K.; Caruso F. Overcoming the Blood-Brain Barrier: The Role of Nanomaterials in Treating Neurological Diseases. Adv. Mater. 2018, 30, e1801362 10.1002/adma.201801362. [DOI] [PubMed] [Google Scholar]
- Wittkowski K. M. A novel oral formulation (CoM pat. pend.) of HP-alpha-cyclodextrin as a safe (not ototoxic) and convenient (oral) drug against Alzheimer’s disease. Alzheimer. Dement. 2022, 18, e066684 10.1002/alz.066684. [DOI] [Google Scholar]
- Wang H.; Xu X.; Pan Y. C.; Yan Y.; Hu X. Y.; Chen R.; Ravoo B. J.; Guo D. S.; Zhang T. Recognition and Removal of Amyloid-β by a Heteromultivalent Macrocyclic Coassembly: A Potential Strategy for the Treatment of Alzheimer’s Disease. Adv. Mater. 2021, 33, e2006483 10.1002/adma.202006483. [DOI] [PubMed] [Google Scholar]
- Lee H. H.; Choi T. S.; Lee S. J.; Lee J. W.; Park J.; Ko Y. H.; Kim W. J.; Kim K.; Kim H. I. Supramolecular inhibition of amyloid fibrillation by cucurbit[7]uril. Angew. Chem., Int. Ed. 2014, 53, 7461. 10.1002/anie.201402496. [DOI] [PubMed] [Google Scholar]
- de Almeida N. E. C.; Do T. D.; Tro M.; LaPointe N. E.; Feinstein S. C.; Shea J.-E.; Bowers M. T. Opposing Effects of Cucurbit[7]uril and 1,2,3,4,6-Penta-O-galloyl-β-d-glucopyranose on Amyloid β25–35 Assembly. ACS Chem. Neurosci. 2016, 7, 218. 10.1021/acschemneuro.5b00280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian Y.; Zhang X.; Li Y.; Shoup T. M.; Teng X.; Elmaleh D. R.; Moore A.; Ran C. Crown ethers attenuate aggregation of amyloid beta of Alzheimer’s disease. Chem. Commun. 2014, 50, 15792. 10.1039/C4CC06029F. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yokoyama T.; Mizuguchi M. Crown Ethers as Transthyretin Amyloidogenesis Inhibitors. J. Med. Chem. 2019, 62, 2076. 10.1021/acs.jmedchem.8b01700. [DOI] [PubMed] [Google Scholar]
- Liu P.; Zhang T.; Chen Q.; Li C.; Chu Y.; Guo Q.; Zhang Y.; Zhou W.; Chen H.; Zhou Z.; Wang Y.; Zhao Z.; Luo Y.; Li X.; Song H.; Su B.; Li C.; Sun T.; Jiang C. Biomimetic Dendrimer-Peptide Conjugates for Early Multi-Target Therapy of Alzheimer’s Disease by Inflammatory Microenvironment Modulation. Adv. Mater. 2021, 33, e2100746 10.1002/adma.202100746. [DOI] [PubMed] [Google Scholar]
- Qu D. H.; Wang Q. C.; Zhang Q. W.; Ma X.; Tian H. Photoresponsive Host-Guest Functional Systems. Chem. Rev. 2015, 115, 7543. 10.1021/cr5006342. [DOI] [PubMed] [Google Scholar]
- Webber M. J.; Langer R. Drug delivery by supramolecular design. Chem. Soc. Rev. 2017, 46, 6600. 10.1039/C7CS00391A. [DOI] [PubMed] [Google Scholar]
- Qi Z.; Schalley C. A. Exploring macrocycles in functional supramolecular gels: from stimuli responsiveness to systems chemistry. Acc. Chem. Res. 2014, 47, 2222. 10.1021/ar500193z. [DOI] [PubMed] [Google Scholar]
- Lou X. Y.; Yang Y. W. Pillar[n]arene-Based Supramolecular Switches in Solution and on Surfaces. Adv. Mater. 2020, 32, e2003263 10.1002/adma.202003263. [DOI] [PubMed] [Google Scholar]
- Braegelman A. S.; Webber M. J. Integrating Stimuli-Responsive Properties in Host-Guest Supramolecular Drug Delivery Systems. Theranostics 2019, 9, 3017. 10.7150/thno.31913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z.; Sun C.; Yang K.; Chen X.; Wang R. Cucurbituril-Based Supramolecular Polymers for Biomedical Applications. Angew. Chem., Int. Ed. 2022, 61, e202206763 10.1002/anie.202206763. [DOI] [PubMed] [Google Scholar]
- Gangemi C. M.A.; Puglisi R.; Pappalardo A.; Trusso Sfrazzetto G. Supramolecular complexes for nanomedicine. Bioorg. Med. Chem. Lett. 2018, 28, 3290. 10.1016/j.bmcl.2018.09.015. [DOI] [PubMed] [Google Scholar]
- Karim A. A.; Dou Q.; Li Z.; Loh X. J. Emerging Supramolecular Therapeutic Carriers Based on Host-Guest Interactions. Chem.-Asian J. 2016, 11, 1300. 10.1002/asia.201501434. [DOI] [PubMed] [Google Scholar]
- Kiss T.; Fenyvesi F.; Bacskay I.; Varadi J.; Fenyvesi E.; Ivanyi R.; Szente L.; Tosaki A.; Vecsernyes M. Evaluation of the cytotoxicity of beta-cyclodextrin derivatives: evidence for the role of cholesterol extraction. Eur. J. Pharm. Sci. 2010, 40, 376. 10.1016/j.ejps.2010.04.014. [DOI] [PubMed] [Google Scholar]
- Crunkhorn S. Crossing the blood-brain barrier. Nat. Rev. Drug Discovery 2021, 20, 740. 10.1038/d41573-021-00151-2. [DOI] [PubMed] [Google Scholar]
- Mukherjee S.; Madamsetty V. S.; Bhattacharya D.; Roy Chowdhury S.; Paul M. K.; Mukherjee A. Recent Advancements of Nanomedicine in Neurodegenerative Disorders Theranostics. Adv. Funct. Mater. 2020, 30, 2003054 10.1002/adfm.202003054. [DOI] [Google Scholar]
- Wu W. H.; Guo J.; Zhang L.; Zhang W. B.; Gao W. Peptide/protein-based macrocycles: from biological synthesis to biomedical applications. RSC. Chem. Biol. 2022, 3, 815. 10.1039/D1CB00246E. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo Q.; Hou C.; Bai Y.; Wang R.; Liu J. Protein Assembly: Versatile Approaches to Construct Highly Ordered Nanostructures. Chem. Rev. 2016, 116, 13571. 10.1021/acs.chemrev.6b00228. [DOI] [PubMed] [Google Scholar]
- Zhu J.; Avakyan N.; Kakkis A.; Hoffnagle A. M.; Han K.; Li Y.; Zhang Z.; Choi T. S.; Na Y.; Yu C.-J.; Tezcan F. A. Protein Assembly by Design. Chem. Rev. 2021, 121, 13701. 10.1021/acs.chemrev.1c00308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hiller S.; Garces R. G.; Malia T. J.; Orekhov V. Y.; Colombini M.; Wagner G. Science 2008, 321, 1206. 10.1126/science.1161302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pollet J.; Chen W. H.; Strych U. Recombinant protein vaccines, a proven approach against coronavirus pandemics. Adv. Drug. Delivery Rev. 2021, 170, 71. 10.1016/j.addr.2021.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong Z.; Luo Q.; Liu J. Artificial enzymes based on supramolecular scaffolds. Chem. Soc. Rev. 2012, 41, 7890. 10.1039/c2cs35207a. [DOI] [PubMed] [Google Scholar]
- Huang X.; Liu X.; Luo Q.; Liu J.; Shen J. Artificial selenoenzymes: designed and redesigned. Chem. Soc. Rev. 2011, 40, 1171. 10.1039/C0CS00046A. [DOI] [PubMed] [Google Scholar]
- Yu X.; Weng Z.; Zhao Z.; Xu J.; Qi Z.; Liu J. Assembly of Protein Cages for Drug Delivery. Pharmaceutics 2022, 14, 2609. 10.3390/pharmaceutics14122609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Csizmar C. M.; Petersburg J. R.; Perry T. J.; Rozumalski L.; Hackel B. J.; Wagner C. R. Multivalent Ligand Binding to Cell Membrane Antigens: Defining the Interplay of Affinity, Valency, and Expression Density. J. Am. Chem. Soc. 2019, 141, 251. 10.1021/jacs.8b09198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y.; Yamada K.; Liddelow S. A.; Smith S. T.; Zhao L.; Luo W.; Tsai R. M.; Spina S.; Grinberg L. T.; Rojas J. C.; Gallardo G.; Wang K.; Roh J.; Robinson G.; Finn M. B.; Jiang H.; Sullivan P. M.; Baufeld C.; Wood M. W.; Sutphen C.; McCue L.; Xiong C.; Del-Aguila J. L.; Morris J. C.; Cruchaga C.; Fagan A. M.; Miller B. L.; Boxer A. L.; Seeley W. W.; Butovsky O.; Barres B. A.; Paul S. M.; Holtzman D. M. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 2017, 549, 523. 10.1038/nature24016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. 10.1111/j.1749-6632.2000.tb06365.x. [DOI] [PubMed] [Google Scholar]
- Song Q.; Huang M.; Yao L.; Wang X.; Gu X.; Chen J.; Chen J.; Huang J.; Hu Q.; Kang T.; Rong Z.; Qi H.; Zheng G.; Chen H.; Gao X. Lipoprotein-based nanoparticles rescue the memory loss of mice with Alzheimer’s disease by accelerating the clearance of amyloid-beta. ACS Nano 2014, 8, 2345. 10.1021/nn4058215. [DOI] [PubMed] [Google Scholar]
- Zhang H.; Jiang W.; Zhao Y.; Song T.; Xi Y.; Han G.; Jin Y.; Song M.; Bai K.; Zhou J.; Ding Y. Lipoprotein-Inspired Nanoscavenger for the Three-Pronged Modulation of Microglia-Derived Neuroinflammation in Alzheimer’s Disease Therapy. Nano Lett. 2022, 22, 2450. 10.1021/acs.nanolett.2c00191. [DOI] [PubMed] [Google Scholar]
- Zhang H.; Zhao Y.; Yu M.; Zhao Z.; Liu P.; Cheng H.; Ji Y.; Jin Y.; Sun B.; Zhou J.; Ding Y. J. Control. Reassembly of native components with donepezil to execute dual-missions in Alzheimer’s disease therapy. Release 2019, 296, 14. 10.1016/j.jconrel.2019.01.008. [DOI] [PubMed] [Google Scholar]
- Denisov I. G.; Sligar S. G. Nanodiscs for structural and functional studies of membrane proteins. Nat. Struct. Mol. Biol. 2016, 23, 481. 10.1038/nsmb.3195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denisov I. G.; Sligar S. G. Nanodiscs in Membrane Biochemistry and Biophysics. Chem. Rev. 2017, 117, 4669. 10.1021/acs.chemrev.6b00690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge Y.; Draycheva A.; Bornemann T.; Rodnina M. V.; Wintermeyer W. Lateral opening of the bacterial translocon on ribosome binding and signal peptide insertion. Nat. Commun. 2014, 5, 5263. 10.1038/ncomms6263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuai R.; Li D.; Chen Y. E.; Moon J. J.; Schwendeman A. High-Density Lipoproteins: Nature’s Multifunctional Nanoparticles. ACS Nano 2016, 10, 3015. 10.1021/acsnano.5b07522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He Y.; Song H. D.; Anantharamaiah G. M.; Palgunachari M. N.; Bornfeldt K. E.; Segrest J. P.; Heinecke J. W. Apolipoprotein A1 Forms 5/5 and 5/4 Antiparallel Dimers in Human High-density Lipoprotein. Mol. Cell. Proteom. 2019, 18, 854. 10.1074/mcp.RA118.000878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Endres K. Apolipoprotein A1, the neglected relative of Apolipoprotein E and its potential role in Alzheimer’s disease. Neural. Regen. Res. 2021, 16, 2141. 10.4103/1673-5374.310669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cochran B. J.; Ong K. L.; Manandhar B.; Rye K. A. APOA1: a Protein with Multiple Therapeutic Functions. Curr. Atheroscler Rep. 2021, 23, 11. 10.1007/s11883-021-00906-7. [DOI] [PubMed] [Google Scholar]
- Palczewski K.; Kumasaka T.; Hori T.; Behnke C. A.; Motoshima H.; Fox B. A.; Trong I. Le; Teller D. C.; Okada T.; Stenkamp R. E.; Yamamoto M.; Miyano M. Crystal structure of rhodopsin: A G protein-coupled receptor. Science 2000, 289, 739. 10.1126/science.289.5480.739. [DOI] [PubMed] [Google Scholar]
- Aurora R.; Rosee G. D. Helix capping. Protein Sci. 1998, 7, 21. 10.1002/pro.5560070103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge Y.; Shen X.; Cao H.; Hao Y.; Jin L.; Shang J.; Wang Y.; Pan T.; Qi Z. A supramolecular hydrophobic guest transport system based on a biological macrocycle. RSC Adv. 2019, 9, 38195. 10.1039/C9RA07054K. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge Y.; Shen X.; Cao H.; Jin L.; Shang J.; Wang Y.; Pan T.; Yang Y.; Qi Z. Biological Macrocycle: Supramolecular Hydrophobic Guest Transport System Based on Nanodiscs with Photodynamic Activity. Langmuir 2019, 35, 7824. 10.1021/acs.langmuir.9b00126. [DOI] [PubMed] [Google Scholar]
- Antina E.; Bumagina N.; Marfin Y.; Guseva G.; Nikitina L.; Sbytov D.; Telegin F. BODIPY Conjugates as Functional Compounds for Medical Diagnostics and Treatment. Molecules 2022, 27, 1396. 10.3390/molecules27041396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J.; Gu B. J.; Masters C. L.; Wang Y. J. A systemic view of Alzheimer disease - insights from amyloid-β metabolism beyond the brain. Nat. Rev. Neurol. 2017, 13, 612. 10.1038/nrneurol.2017.111. [DOI] [PubMed] [Google Scholar]
- Song Q.; Song H.; Xu J.; Huang J.; Hu M.; Gu X.; Chen J.; Zheng G.; Chen H.; Gao X. Biomimetic ApoE-Reconstituted High Density Lipoprotein Nanocarrier for Blood-Brain Barrier Penetration and Amyloid Beta-Targeting Drug Delivery. Mol. Pharmaceutics 2016, 13, 3976. 10.1021/acs.molpharmaceut.6b00781. [DOI] [PubMed] [Google Scholar]
- Hamley I. W. The amyloid beta peptide: a chemist’s perspective. Role in Alzheimer’s and fibrillization. Chem. Rev. 2012, 112, 5147. 10.1021/cr3000994. [DOI] [PubMed] [Google Scholar]
- Pascoal T. A.; Benedet A. L.; Ashton N. J.; Kang M. S.; Therriault J.; Chamoun M.; Savard M.; Lussier F. Z.; Tissot C.; Karikari T. K.; Ottoy J.; Mathotaarachchi S.; Stevenson J.; Massarweh G.; Scholl M.; de Leon M. J.; Soucy J. P.; Edison P.; Blennow K.; Zetterberg H.; Gauthier S.; Rosa-Neto P. Microglial activation and tau propagate jointly actross braak stages. Nat. Med. 2021, 27, 1592. 10.1038/s41591-021-01456-w. [DOI] [PubMed] [Google Scholar]
- Mandal P. K.; Pettegrew J. W. Alzheimer’s disease: soluble oligomeric Abeta(1–40) peptide in membrane mimic environment from solution NMR and circular dichroism studies. Neurochem. Res. 2004, 29, 2267. 10.1007/s11064-004-7035-1. [DOI] [PubMed] [Google Scholar]
- LaFerla F. M.; Green K. N.; Oddo S. Intracellular amyloid-beta in Alzheimer’s disease. Nat. Rev. Neurosci. 2007, 8, 499. 10.1038/nrn2168. [DOI] [PubMed] [Google Scholar]
- Chen X.; Firulyova M.; Manis M.; Herz J.; Smirnov I.; Aladyeva E.; Wang C.; Bao X.; Finn M. B.; Hu H.; Shchukina I.; Kim M. W.; Yuede C. M.; Kipnis J.; Artyomov M. N.; Ulrich J. D.; Holtzman D. M. Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature 2023, 615, 668. 10.1038/s41586-023-05788-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



