SDRP Journal of Nanotechnology & Material Science

ISSN: 2574-1888

Impact Factor: 0.621

VOLUME: 2 ISSUE: 1

Page No: 94-105

NANOTHERAPY TO DELAY COGNITIVE IMPAIRMENT: USING COLLOIDAL NANOCARRIERS TO BLOCK AMYLOID-β-INDUCED DAMAGE IN BRAIN CELL MEMBRANES


Corresponding Author

Joseph S. D'Arrigo, Ph.D

 Cavitation-Control Technology Inc., Farmington, CT 06032, USA

Email: cavcon@ntplx.net

Citation

Joseph S. D'Arrigo, NANOTHERAPY TO DELAY COGNITIVE IMPAIRMENT: USING COLLOIDAL NANOCARRIERS TO BLOCK AMYLOID-β-INDUCED DAMAGE IN BRAIN CELL MEMBRANES(2019)SDRP Journal of Nanotechnology & Material Science 2(1)

Abstract

Background: Numerous published studies indicate that microvascular endothelial dysfunction precedes cognitive decline in Alzheimer's disease, and that preservation of a healthy cerebrovascular endothelium can be an important therapeutic target.

Methods: By incorporating appropriate drug(s) into biomimetic (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic which targets certain cell-surface scavenger receptor, mainly class B type I (i.e., SR-BI), and crosses the blood-brain barrier. Documented similarities in lipid composition between high-density lipoproteins (HDL) and the biomimetic (nanoemulsion) nanocarrier particles can partially  simulate or mimic the known heterogeneity (i.e., subpopulations or subspecies) of HDL particles. Moreover, the above-described type BI scavenger receptor (i.e., either SR-BI (rodent) and/or CLA-1 (human) orthologs) has been shown to be a multifunctional receptor able to bind a broad variety of ligands, including HDL and chylomicron remnants.

Results: Such colloidal-nanocarrier targeting allows for various Alzheimer's-related cell types to be simultaneously searched out, in vivo, for localized drug treatment. Using various lipids and their mixtures to form self-assembled non-lamellar nanostructures, it has continually been reported possible to successfully obtain stable colloidal dispersions of (liquid-crystalline) lipid cubic phases with well-defined particle size and morphology. In particular, within the range of self-assembled phases in model surfactant-like lipid systems, various investigators further emphasize that monglyceride-based lyotropic liquid-crystalline phases are relatively unique owing to their rich polymorphism in water and potential application as drug nanocarriers.

Conclusion: This (colloidal-nanocarrier) in vivo targeting advantage may be particularly important for repurposing an FDA-approved drug, especially one which has shown the added ability to restore some cognitive functions in certain animals models of Alzheimer's disease (e.g., the anticancer drug bexarotene). Bexarotene (and several analogs or other candidate-repurposing drugs) up to now, by itself (i.e., without nanocarrier), displayed poor CNS penetration in human subjects.

Keywords:  Alzheimer's disease, blood-brain barrier, cognitive aging, dementia, drug targeting, lipid cubic phases, nanoemulsion, scavenger receptors, SR-BI

Introduction

Background

A frequent co-morbidity of cerebrovascular pathology and Alzheimer's disease pathology has been observed over past decades. Accordingly, much evidence has been reported which indicates that microvascular endothelial dysfunction, due to cerebrovascular risk factors (e.g., atherosclerosis, obesity, diabetes, smoking, hypertension, aging), precedes cognitive decline in Alzheimer's disease and contributes to its pathogenesis. It is no surprise, therefore, that vascular brain lesions are very common in people over 70 years old, and recent reviews (e.g., [1,2]) provide much evidence that a large proportion of dementia cases may be attributable to cerebrovascular disease. As a result, vascular cognitive impairment and dementia (VCID) is the second leading cause of dementia behind Alzheimer's disease [4,10,11].

 

Endothelial Dysfunction as a Therapeutic Target for Cognitive Impairment

It has been reconfirmed in the current literature (see below) that receptor-mediated endocytosis/transcytosis via lipoprotein receptors, particularly scavenger receptors (including class B type I, i.e., SR-BI), remains a major route for drug delivery across the blood-brain barrier (BBB). Accordingly, endothelial-cell modulation and repair is feasible by pharmacological targeting [1,2,12-26] via SR-BI receptors (cf. [27-31]). Recently, Fung et al. [32] specifically reported that SR-BI mediates the uptake and transcytosis of HDL across brain microvascular endothelial cells (i.e., across the BBB). Since SR-BI has already been identified as a major receptor for HDL (with their major apolipoprotein (apo)A-I) as well as for the recently reviewed [1,2] “lipid-coated microbubble/nanoparticle-derived” (LCM/ND) nanoemulsion (see below), this multitasking lipid nanoemulsion can arguably serve as a targeted, apoA-I-based, (SR-BI mediated) therapeutic agent for common (late-onset) dementias [2,29,31,33-37] (cf. [38-43]).

This targeted-drug-delivery approach, using the proposed LCM/ND lipid nanoemulsion for treating the more common (late-onset) dementias, receives added impetus from continual findings of cerebrovascular pathology [1,44–51] and an apparent endothelium dysfunction [2,28–36,52–61] in both Alzheimer’s disease and its major risk factors [1,2,62-71]. Furthermore, this (intravenous) combination therapeutic would make it possible for various cell types, all potentially implicated in Alzheimer’s disease (see [1,2] for reviews; cf. [72,73]), to be simultaneously sought out and better reached for localized drug treatment of brain tissue in vivo [73] (cf. [74]).

Consistent with the above considerations concerning endothelial dysfunction in Alzheimer's disease, ApoA-I has very recently been reported by Camacho et al. [75] to play a special role in cerebral amyloid angiopathy (CAA). CAA is observed in more than 90% of patients with Alzheimer's disease, although it additionally has an independent contribution to the cognitive deterioration associated with age. CAA can be neuropathologically classified as CAA  type I or type II, where type I is characterized by amyloid-β deposition in cerebral capillaries (versus type II where cerebral capillaries are not involved). The human data (from autopsy brains of 20 post-mortem cases), reported by Camacho et al., revealed that the ApoA-I immunohistochemical staining was localized strongly toward capillary walls with CAA (i.e., especially toward CAA type I pathology) [75].

Also, as alluded to above (see Abstract), the previously documented similarities in lipid composition among HDL (as well as native low-density lipoproteins (LDL) and modified LDL) and LCM/ND nanoemulsion particles can partially simulate or mimic the known heterogeneity (i.e., subpopulations or subspecies) of HDL particles (see [73] for a review). Moreover, the above-described type BI scavenger receptor (i.e., either SR-BI (rodent) and/or CLA-1 (human) orthologs) has been shown to be a multifunctional receptor able to bind a broad variety of ligands, including HDL, LDL, oxidized LDL (OxLDL), very-low-density lipoproteins (VLDL), and chylomicron remnants. The presence of amphipathic helices is a common feature of “exchangeable apolipoproteins”, which are known to be the primary ligands (including notably apoA-I) for SR-BI [3]. In view of all the above considerations, SR-BI emerges as the most plausible candidate (of all lipoprotein receptors) for major involvement in the enhanced endocytosis of LCM/ND nanoemulsion(s) for targeted drug delivery [73].

 

LCM/ND Nanoemulsion Type, Lipid Cubic Phases, and Biomimetic Nanocarriers

The self-assembling LCM/ND lipid nanoemulsion class comprises nonionic lipids exclusively (e.g., [76]) throughout its coated microbubble's and/or related nanoparticle's (i.e., related lipid polymorphs') supramolecular structures(s). This biobased lipid composition of LCM/ND nanoemulsions (i.e., comprising glycerides and cholesterol compounds) is similar to lipids contained in several types of plama lipoproteins; accordingly, when these LCM/ND nanoemulsion particles are injected into the bloodstream, they likely acquire (i.e., bind) plasma apolipoprotein(s) – including notably apoA-I [73].

Importantly, monoglyceride is the largest single-lipid fraction (by wt. %) of the powdered solid lipid surfactants used to produce the (Filmix®) LCM/ND nanoemulsions [73]. As a group, monoglycerides exhibit different phase behaviors when they are exposed to water [77] (cf. [78-81]). In agreement with numerous other investigators, Kaasgaard and Drummond [82] also state that all these types of liquid-crystalline phases are frequently stable in excess water, which facilitates the preparation of nanoparticle dispersions and makes them suitable candidates for the encapsulation and controlled release of drugs (cf. [83-89]).

In particular, the self-assembly of varied and useful dispersed cubic phases (among other liquid-crystalline phases) depends heavily on the acyl chain length of the glycerides (primarily monoglycerides) placed in contact with water [73]. There is great interest to utilize these dispersed cubic phases for the administration of drugs, or for the formulation of new delivery systems [73,87]. The (lyotropic or solvent-induced) cubic liquid-crystalline phases may be classified into two distinct classes: bicontinuous cubic phases [90-94] and micellar or discontinuous (e.g., type Fd3m) cubic phases [89,90]. As reviewed by Garg et al. [77], monoglycerides spontaneously form bicontinuous cubic phases upon the addition of water, are relatively insoluble (allowing the formation of colloidal dispersions of cubic phases) and are resistant to changes in temperature. Accordingly, lipid nanoparticles comprising interior liquid-crystalline structures of curved lipid membranes (i.e., dispersed cubic phases) have been used to solubilize, encapsulate, and deliver medications to disease areas within the body [77] (see also [73,95-105]).

In addition to the above-described category of various bicontinuous cubic phases, the other above-named category referred to as “micellar or discontinuous” cubic phases are also worthy of comment at this point. Of particular interest within this latter category is the well-studied micellar cubic structure of the type Fd3m (which is often denoted by the number Q227) (e.g., [89]). Luzzati and coworkers have reported that this Fd3m cubic phase evidently requires a heterogeneous mixture of polar lipids [91]: Using the lipid examples they cite (and the lipid classification system of Small [81]), this Fd3m phase apparently must include both at least one (sufficiently polar) insoluble swelling amphiphilic lipid (e.g., monoglyceride ) and at least one (weakly polar) insoluble nonswelling amphiphilic lipid (e.g., diglyceride and/or cholesterol) (e.g., [3]; cf. [73]) in order to self-assemble properly in (excess) water. Hence, the dispersed Fd3m cubic phase can represent a lipid/water system which is particularly relevant to the earlier-described (Filmix®) LCM/ND lipid nanoemulsion formulation(s) on account of the fact that the patent claims describing the precise lipid composition of such nanoemulsion formulations (see especially Claim #1 in [76]) specifically include cholesterol and three categories of (saturated) glycerides, that is, tri-, di-, and monoglycerides (see [76]). In view of all the advantageous attributes of monoglycerides (recounted in the preceding paragraphs), and since (saturated) monoglyceride represents the largest single-lipid fraction of the LCM/ND lipid nanoemulsion type, the monoglyceride content probably plays a dominant role in supporting the evident long-term stability of the liquid-crystalline lipid nanoparticles in such nanoemulsions (see also [73] for a detailed review).

Besides certain glyceride-based liquid-crystalline systems displaying colloidal stability in excess water, the same important attribute has been documented for cholesterol and cholesterol esters – all of which are present in LCM/ND nanoemulsion formulations [73]. For example, cholesterol and its esters change the packing structure of lipids, and in high

concentrations they are known to induce the formation of a liquid-crystal phase [106]. In addition, Kuntsche et al. [107,108] have prepared lipid nanoparticles in the (mesomorphic or) liquid-crystalline phase from cholesterol esters with saturated acyl chains. In accord with the above observations and considerations, the substantial concentrations of cholesterol esters and cholesterol in the LCM/ND nanoemulsion formulation likely further contribute to the known long-term stability of this nanoemulsion's (liquid-crystalline) lipid nanoparticles in excess water, thereby providing a persistent carrier matrix upon exposure to liquids such as blood plasma [73].

To conclude, self-assembled (colloidal mesophase) lipid nanoemulsions (e.g., [92-98]), particularly those predominantly containing dispersed cubic-phase lipid nanoparticles (e.g., [3,73,99-105]), continue to receive growing attention in pharmaceutical and/or biological fields. The main reason behind much of this attention is the fact that nonlamellar lipid nanostructures, such as cubic liquid-crystalline phases, have wide potential as delivery systems for numerous drugs, cosmetics, and food applications (cf. [3]). A recurring example of a largely monoglyceride-based drug-delivery agent category is the multitasking LCM/ND nanoemulsion formulation (cf. above). In this particular targeted-delivery approach, the self-assembled “lipid particle” structure itself (upon intravenous injection of the LCM/ND nanoemulsion) is apparently successfully utilized as the “active” targeting ligand – which is directed via (adsorption of) plasma lipoproteins toward the appropriate receptors on the target-cell surface. These dispersed liquid-crystalline lipid particles, of the LCM/ND nanoemulsion formulation, are colloidally stable nanocarriers which very likely represent liquid-crystalline inverse-topology nanotransporters (nanocarriers), i.e., dispersed lipid cubic phases (cf. [73]).

 

Amyloid-β Ion Channel Hypothesis of Alzheimer's Disease

As explained in many reviews (e.g., [109,110]) by different investigators, it has been recognized for over two decades that disturbance of the intracellular calcium homeostasis is central to the pathophysiology of several neurodegenerative disorders. As concerns Alzheimer's disease, it is believed by many researchers that enhanced calcium load may be brought about by extracellular accumulation of amyloid-β (Aβ) in the brain. Such studies have laid the foundation for the popular idea that amyloid-β peptides (39-42 amino acid molecules) are, in part, toxic to brain tissue because they form aberrant ion channels in cellular membranes and thereby disrupt Ca2+ homeostasis in brain tissue and increase intracellular Ca2+. More specifically, later studies indicated that soluble forms of Aβ facilitate influx through calcium-conducting ion channels in the plasma membrane, leading to excitotoxic neurodegeneration [109,110].

Historical support for the above amyloid-β ion channel hypothesis, or so-called “calcium hypothesis” has also been observed at the clinical level [111]. Namely, there is little correlation between the amounts of fibrillar (insoluble) deposit at autopsy and the clinical severity of Alzheimer's disease. In contrast, a good correlation exists between early cognitive impairment and levels of soluble forms of Aβ in the brain [112]. (Aggregation of Aβ proceeds from formation of soluble (low molecular weight) spherical oligomers toward eventually assuming a final and stable conformation as insoluble fibrils from which amyloid-β plaques are constituted. Neurotoxicity is associated with soluble aggregates (i.e., oligomers) of Aβ rather than with the plaques themselves.) Accordingly, related experimental work has already shown that application of soluble Aβ oligomers (but not monomers or fibrils) to cultured neuroblastoma cells evoked large increases in cytosolic calcium that arise largely through Ca2+ influx across the plasma membrane [112].

As summarized by Di Scala et al. [111], the structure of amyloid pores has been extensively studied by ultrastructural methods. In particular, one group of investigators recently applied strategies (widely used to examine the structure of membrane proteins) to study the two major Aβ variants, namely, Aβ(1-40) and Aβ(1-42).  Under the optimized detergent micelle conditions: 1) Aβ(1-40) aggregated into amyloid fibrils; 2) contrariwise, Aβ(1-42) assembled into oligomers that inserted into lipid bilayers as well-defined pores [113]. (These amyloid pores adopted characteristics of a β-barrel arrangement.) Because Aβ(1-42), relative to Aβ(1-40), has a more prominent role in Alzheimer's disease, the higher propensity of Aβ(1-42) to form β-barrel pore-forming oligomers is an indication of their importance in Alzheimer's disease [113]. Very recently, a different research group reported very similar findings [114]. As background for their study, these latter authors point out that: elevated Aβ(1-42) plasma levels have been correlated with the progression of late-onset forms of Alzheimer's disease; Aβ(1-42) is significantly more neurotoxic than Aβ(1-40) both in vivo and in neuronal cell culture; and memory impairment is believed to be driven by Aβ(1-42) disruption of long-term (hippocampal) potentiation. In accordance with these considerations, the authors' own detailed experimental data [114] indicated that Aβ(1-42) assemblies in oligomeric preparations form ion channels (in membranes excised from cells of neuronal origin). In contrast, Aβ(1-40) oligomers, fibrils, and monomers did not form channels. Moreover, ion-channel-conductance results suggested that Aβ(1-42) oligomers, but not monomers and fibrils, formed pore structures. The authors concluded that their findings demonstrate that only Aβ(1-42) contains unique structural features that facilitate membrane insertion and channel formation, now aligning ion channel formation with the neurotoxic effect of Aβ(1-42) compared to Aβ(1-40) in Alzheimer's disease [114].

 

Promise of Bexarotene (or analogs) to Inhibit Cognitive Decline in Humans

The preceding discussion of amyloid pore formation, in the cellular membranes of brain tissue, leads to another important consideration – the role of cholesterol. Namely, cholesterol is required for the assembly of amyloid pores formed by Aβ(1-42) [111]. Therefore, an amphipathic drug (such as bexarotene) which competes with cholesterol for binding to Aβ(1-42) would be capable of preventing oligomeric channel formation (at least in vitro). Such a strategy has already been contemplated for the treatment of Alzheimer's and other neurodegenerative diseases that involve cholesterol-dependent toxic oligomers [115]. However, when oral administration of bexarotene was employed subsequently in a Phase Ib (proof of mechanism) clinical trial [116], bexarotene displayed poor CNS penetration in normal human subjects. (Hence, the observed absence of an effect on Aβ metabolism was likely reflective of the low CNS-levels of bexarotene achieved [116](cf. [117])).

Nonetheless, at least two recently published reports (both in 2017) on bexarotene indicate a continuing interest in the ability of this FDA-approved (anticancer) drug to: 1) bind free Aβ peptide, as well as 2) bexarotene's previously reported positive effects in Alzheimer's-disease mouse models [118,119] (cf. [120,121]). Such past studies in animal models of Alzheimer's disease, concerning the beneficial effects of bexarotene, have also motivated a detailed analysis by Fantini et al. [122] to elucidate the mechanisms underlying the anti-Alzheimer properties of bexarotene in brain cells. These investigators demonstrated that bexarotene shares structural analogy with cholesterol: both bexarotene and cholesterol are amphipathic compounds, with a large apolar part consisting of a succession of hydrocarbon rings and a small polar headgroup (hydroxyl for cholesterol, carboxylate for bexarotene). Because it is the first drug that can both inhibit the binding of cholesterol to Aβ(1-42) and prevent calcium-permeable amyloid pore formation in the plasma membrane of brain cells, bexarotene might be considered as the prototype of a new class of anti-Alzheimer compounds [122]. (Note that because bexarotene shares structural analogy with cholesterol, and the above-described LCM/ND nanoemulsion contains substantial concentrations of cholesterol esters and cholesterol (see above), incorporation of the bexarotene molecule into the LCM/ND nanocarrier is expected to represent an uncomplicated, straightforward formulation procedure commercially.) Moreover, Casali et al. [123] have very recently reported that treatment of an Alzheimer's-disease mouse model with (this FDA-approved anticancer drug) bexarotene resulted in enhanced cogniton in the APP/PS1 mice which resembled earlier findings. Strikingly, the authors observed sustained cognitive improvements in the mice even when bexarotene treatment was discontinued for 2 weeks. Also, they observed a sustained reduction in microgliosis and plaque burden, following drug withdrawal, exclusively in the hippocampus. Casali et al. concluded that bexarotene selectively modifies aspects of neuroinflammation in a region-specific manner to reverse hippocampal-dependent cognitive deficits in Alzheimer's-disease (APP/PS1) mice [123].

Additional molecular aspects, concerning the membrane-related mechanisms for the known neuroprotective effect, of bexarotene action on brain tissue continue to be suggested and/or described in the recent literature (cf. [124,125]). In the most recently published study, Kamp et al. [126] show by NMR and CD spectroscopy that bexarotene directly interacts with the transmembrane domain of the amyloid precursor protein (APP) in a region where cholesterol binds. (Note that Aβ peptides are derived from APP, by the sequential action of β- and γ-secretases. γ-Secretase cleavage occurs in the transmembrane domain, of the C-terminal fragment left by β-secretase cleavage, and results in the release of Aβ peptides of various lengths [126]. The longer, neurotoxic, Aβ(1-42) peptide is highly aggregation prone and represents the major Aβ species deposited in the brain [126-129]. Cholesterol promotes Aβ(1-42) aggregation by enhancing its primary nucleation rate by up to 20-fold  [129].) Kamp et al. argue that their data [126] suggest that bexarotene is a pleiotropic molecule that interferes with Aβ metabolism through multiple mechanisms. More specifically, earlier work by Di Scala et al. [115] provided evidence that bexarotene competed with cholesterol for binding to Aβ and prevented oligomeric channel formation. Di Scala et al. argue that their findings indicate that it is possible to prevent the generation of neurotoxic oligomers by targeting the cholesterol-binding domain of Aβ peptides [115]. Note that such blocking of amyloid-β-induced neurotoxic pore formation can be expected to avoid exacerbation of blood-brain barrier breakdown, already occurring at lower levels in aged humans with cognitive decline [130], and thereby prevent reaching higher levels of BBB breakdown associated with cognitive impairment (and/or eventually dementia) in late-onset Alzheimer's disease [130-132].

Conclusion

By incorporating the appropriate drug(s) into biomimetic (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic which targets certain cell-surface scavenger receptors, mainly class B type I (SR-BI), and crosses the BBB. Such biomimetic-nanoemulsion targeting allows for various Alzheimer's-related cell types to be simultaneously searched out, in vivo, for localized drug treatment. The proposed multitasking nanocarrier therapeutic appears likely to display greater efficacy at different stages of Alzheimer's disease. This (colloidal-nanocarrier) in vivo targeting advantage may be particularly important for repurposing an FDA-approved drug, especially one (such as the anticancer drug bexarotene) which has shown the added ability to restore some cognitive functions in certain animal models of Alzheimer's disease.

Acknowledgement

This research did not receive any specific grant from funding agencies in the public, commercial, or nonprofit sectors.

References

  1. D'Arrigo, J.S. Alzheimer's disease, brain injury, and CNS nanotherapy in humans: Sonoporation augmenting drug targeting. Med. Sci. 2017, 5, 29. PMCid:PMC5753658

    View Article           
  2. D'Arrigo, J.S. Nanotherapy for Alzheimer's disease and vascular dementia: Targeting senile endothelium. Adv. Colloid Interface Sci. 2018, 251, 44-54. PMid:29274774

    View Article      PubMed/NCBI     
  3. D'Arrigo, J.S. Targeting early dementia: Using lipid cubic phase nanocarriers to cross the blood-brain barrier. Biomimetics 2018, 3, 4. PMid:31105226

    View Article      PubMed/NCBI     
  4. Dichgans, M.; Leys, D. Vascular cognitive impairment. Circ. Res. 2017, 120, 573-591. PMid:28154105

    View Article      PubMed/NCBI     
  5. Greenberg, S.M. Vascular disease and neurodegeneration: Advancing together. Lancet Neurol. 2017, 16, 333. 30086-8

    View Article           
  6. Kalaria, R.N. Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer's disease. Acta Neuropathol. 2016, 131, 659-685. PMid:27062261

    View Article      PubMed/NCBI     
  7. Duncombe, J.; Kitamura, A.; Hase, Y.; Ihara, M.; Kalaria, R.N.; Horsburgh, K. Chronic cerebral hypoperfusion: A key mechanism leading to vascular cognitive impairment and dementia. Closing the translational gap between rodentmodels and human vascular cognitive impairment and dementia. Clin. Sci. 2017, 131, 2451-2468. PMid:28963120

    View Article      PubMed/NCBI     
  8. Perrotta, M.; Lembo, G.; Carnevale, D. Hypertension and dementia: Epidemiological and experimental evidence revealing a detrimental relationship. Int. J. Mol. Sci. 2016, 17, 347. PMid:27005613

    View Article      PubMed/NCBI     
  9. Sudduth, T.L.;Weekman, E.M.; Price, B.R.; Gooch, J.L.;Woolums, A.; Norris, C.M.; Wilcock, D.M. Time-course of glial changes in the hyperhomocysteinemia model of vascular cognitive impairment and dementia (VCID). Neuroscience 2017, 341, 42-51. PMid:27890830

    View Article      PubMed/NCBI     
  10. Bhat, N.R. Vasculoprotection as a convergent, multi-targeted mechanism of anti-AD therapeutics and interventions. J. Alzheimers Dis. 2015, 46, 581-591. PMid:26402511

    View Article      PubMed/NCBI     
  11. Alzheimer's Disease International. World Alzheimer Report 2016; Alzheimer's Disease International: London, UK, 2016. Available online: www.alz.co.uk/worldreport2016 (accessed on 20 February 2018).

  12. Srimanee, A.; Regberg, J.; Hallbrink, M.; Vajragupta, O.; Langel, U. Role of scavenger receptors in peptide-based delivery of plasmid DNA across a blood-brain barrier model. Int. J. Pharm. 2016, 500, 128-135. PMid:26773601

    View Article      PubMed/NCBI     
  13. De Boer, A.G.; van der Sandt, I.C.J.; Gaillard, P.J. The role of drug transporters at the blood-brain barrier. Annu. Rev. Pharmacol. Toxicol. 2003, 43, 629-656. PMid:12415123

    View Article      PubMed/NCBI     
  14. Almer, G.; Mangge, H.; Zimmer, A.; Prassl, R. Lipoprotein-related and apolipoprotein-mediated delivery systems for drug targeting and imaging. Curr. Med. Chem. 2015, 22, 3631-3651. PMid:26180001

    View Article      PubMed/NCBI     
  15. Preston, J.E.; Abbott, J.; Begley, D.J. Transcytosis of macromolecules at the blood-brain barrier. Adv. Pharmacol. 2014, 71, 147-163. PMid:25307216

    View Article      PubMed/NCBI     
  16. Di Marco, L.Y.; Venneri, A.; Farkas, E.; Evans, P.C.; Marzo, A.; Frangi, A.F. Vascular dysfunction in the pathogenesis of Alzheimer's disease-A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol. Dis. 2015, 82, 593-606. PMid:26311408

    View Article      PubMed/NCBI     
  17. Salmina, A.B.; Inzhutova, A.I.; Malinovskaya, N.A.; Petrova, M.M. Endothelial dysfunction and repair in Alzheimer-type neurodegeneration: Neuronal and glial control. J. Alzheimers Dis. 2010, 22, 17-36. PMid:20847414

    View Article      PubMed/NCBI     
  18. Tong, X.K.; Hamel, E. Simvastatin restored vascular reactivity, endothelial function and reduced string vessel pathology in a mouse model of cerebrovascular disease. J. Cereb. Blood Flow Metab. 2015, 35, 512-520. PMid:25564230

    View Article      PubMed/NCBI     
  19. Carradori, D.; Gaudin, A.; Brambilla, D.; Andrieux, K. Application of nanomedicine to the CNS diseases. Int. Rev. Neurobiol. 2016, 130, 73-113. PMid:27678175

    View Article      PubMed/NCBI     
  20. Koster, K.P.; Thomas, R.; Morris, A.W.; Tai, L.M. Epidermal growth factor prevents oligomeric amyloid-induced angiogenesis deficits in vitro. J. Cereb. Blood Flow Metab. 2016, 36, 1865-1871. PMid:27634936

    View Article      PubMed/NCBI     
  21. Zenaro, E.; Piacentino, G.; Constantin, G. The blood-brain barrier in Alzheimer's disease. Neurobiol. Dis. 2016, 107, 41-56. PMid:27425887

    View Article      PubMed/NCBI     
  22. Qosa, H.; Mohamed, A.; Al Rihani, S.B.; Batarseha, Y.S.; Duong, Q.V.; Keller, J.N.; Kaddoumi, A. High-throughput screening for identification of blood-brain barrier integrity enhancers: A drug repurposing opportunity to rectify vascular amyloid toxicity. J. Alzheimers Dis. 2016, 53, 1499-1516. PMid:27392852

    View Article      PubMed/NCBI     
  23. Hostenbach, S.; D'haeseleer, M.; Kooijman, R.; De Keyser, J. The pathophysiological role of astrocytic endothelin-1. Prog Neurobiol. 2016, 144, 88-102. PMid:27132521

    View Article      PubMed/NCBI     
  24. Koizumi, K.;Wang, G.; Park, L. Endothelial dysfunction and amyloid--induced neurovascular alterations. Cell. Mol. Neurobiol. 2016, 36, 155-165. PMid:26328781

    View Article      PubMed/NCBI     
  25. Goldwaser, E.L.; Acharya, N.K.; Sarkar, A.; Godsey, G.; Nagele, R.G. Breakdown of the cerebrovasculature and blood-brain barrier: A mechanistic link between diabetes mellitus and Alzheimer's disease. J. Alzheimers Dis. 2016, 54, 445-456. PMid:27497477

    View Article      PubMed/NCBI     
  26. Bredesen, D.E. Reversal of cognitive decline: A novel therapeutic program. Aging (Albany, NY) 2014, 6, 707-717. PMid:25324467

    View Article      PubMed/NCBI     
  27. Mahringer, A.; Reichel, V.; Ott, M.; MacLean, C.; Reimold, I.; Hollnack-Pusch, E.; Fricker, G. Overcoming the blood brain barrier: The challenge of brain drug targeting. J. Nanoneurosci. 2012, 2, 5-19.

    View Article           
  28. Robert, J.; Button, E.B.; Stukas, S.; Boyce, G.K.; Gibbs, E.; Cowan, C.M.; Gilmour, M.; Cheng, W.H.; Soo, S.K.; Yuen, B.; et al. High-density lipoproteins suppress Aβ-induced PBMC adhesion to human endothelial cells in bioengineered vessels and in monoculture. Mol. Neurodegener. 2017, 12, 60. PMid:28830501

    View Article      PubMed/NCBI     
  29. Vishnyakova, T.G.; Bocharov, A.V.; Baranova, I.N.; Chen, Z.; Remaley, A.T.; Csako, G.; Eggerman, T.L.; Patterson, A.P. Binding and internalization of lipopolysaccharide by CLA-1, a human orthologue of rodent scavenger receptor B1. J. Biol. Chem. 2003, 278, 22771-22780. PMid:12651854

    View Article      PubMed/NCBI     
  30. Darlington,D.; Li, S.; Hou, H.; Habib, A.; Tian, J.; Gao, Y.; Ehrhart, J.; Sanberg, P.R.; Sawmiller, D.; Giunta, B.; et al. Human umbilical cord blood-derived monocytes improve cognitive deficits and reduce amyloid-pathology in PSAPP mice. Cell Transplant. 2015, 24, 2237-2250. PMid:26230612

    View Article      PubMed/NCBI     
  31. Chang, E.H.; Rigotti, A.; Huerta, P. Age-related influence of the HDL receptor SR-BI on synaptic plasticity and cognition. Neurobiol. Aging 2009, 30, 407-419. PMid:17719144

    View Article      PubMed/NCBI     
  32. Fung, K.Y.;Wang, C.; Nyegaard, S.; Heit, B.; Fairn, G.D.; Lee,W.L. SR-BI mediated transcytosis of HDL in brain microvascular endothelial cells is independent of caveolin, clathrin, and PDZK1. Front. Physiol. 2017, 8, 841. PMid:29163190

    View Article      PubMed/NCBI     
  33. Robert, J.; Stukas, S.; Button, E.; Cheng, W.H.; Lee, M.; Fan, J.; Wilkinson, A.; Kulic, I.; Wright, S.D.; Wellington, C.L. Reconstituted high-density lipoproteins acutely reduce soluble brain A levels in symptomatic APP/PS1 mice. Biochim. Biophys. Acta 2016, 1862, 1027-1036. PMid:26454209

    View Article      PubMed/NCBI     
  34. Armstrong, S.M.; Sugiyama, M.G.; Fung, K.Y.Y.; Gao, Y.; Wang, C.; Levy, A.S.; Azizi, P.; Roufaiel,M.; Zhu, S.N.; Neculai, D.; et al. A novel assay uncovers an unexpected role for SR-BI in LDL transcytosis. Cardiovasc. Res. 2015, 108, 268-277. PMid:26334034

    View Article      PubMed/NCBI     
  35. Hottman, D.A.; Chernick, D.; Cheng, S.; Wang, Z.; Li, L. HDL and cognition in neurodegenerative disorders. Neurobiol. Dis. 2014, 72, 22-36. PMid:25131449

    View Article      PubMed/NCBI     
  36. Velagapudi, S.; Yalcinkaya, M.; Piemontese, A.; Meier, R.; Norrelykke, S.F.; Perisa, D.; Rzepiela, A.; Stebler, M.; Stoma, S.; Zanoni, P.; et al. VEGF-A regulates cellular localization of SR-BI as well as transendothelial transport of HDL but not LDL. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 794-803. PMid:28360088

    View Article      PubMed/NCBI     
  37. Choi, H.J.; Seo, E.H.; Yi, D.; Sohn, B.K.; Choe, Y.M.; Byun, M.S.; Lee, J.M.; Woo, J.I.; Lee, D.Y.Amyloid-independent amnestic mild cognitive impairment and serum apolipoprotein A1 levels. Am. J. Geriatr. Psychiatry 2016, 24, 144-153. PMid:26238231

    View Article      PubMed/NCBI     
  38. Kitamura, Y.; Usami, R.; Ichihara, S.; Kida, H.; Satoh, M.; Tomimoto, H.; Murata, M.; Oikawa, S. Plasma protein profiling for potential biomarkers in the early diagnosis of Alzheimer's disease. Neurol. Res. 2017, 39, 231-238. PMid:28107809

    View Article      PubMed/NCBI     
  39. Lazarus, J.; Mather, K.A.; Armstrong, N.J.; Song, F.; Poljak, A.; Thalamuthu, A.; Lee, T.; Kochan, N.A.; Brodaty, H.; Wright, M.J.; et al. DNA methylation in the apolipoprotein-A1 gene is associated with episodic memory performance on healthy older individuals. J. Alzheimers Dis. 2015, 44, 175-182. PMid:25261444

    View Article      PubMed/NCBI     
  40. Ma, C.; Li, J.; Bao, Z.; Ruan, Q.; Yu, Z. Serum levels of apoA1 and apoA2 are associated with cognitive status in older men. Biomed. Res. Int. 2015, 2015, 481621. PMid:26682220

    View Article      PubMed/NCBI     
  41. Slot, R.E.; Van Harten, A.C.; Kester, M.I.; Jongbloed, W.; Bouwman, F.H.; Teunissen, C.E.; Scheltens, P.; Veerhuis, R.; van der Flier,W.M. Apolipoprotein A1 in cerebrospinal fluid and plasma and progression to Alzheimer's disease in non-demented elderly. J. Alzheimers Dis. 2017, 56, 687-697. PMid:28035918

    View Article      PubMed/NCBI     
  42. Yin, Z.G.; Li, L.; Cui, M.; Zhou, S.M.; Yu, M.M.; Zhou, H.D. Inverse relationship between apolipoprotein A-I and cerebral white matter lesions: A cross-sectional study in middle-aged and elderly subjects. PLoS ONE 2014, 9, e97113. PMid:24820970

    View Article      PubMed/NCBI     
  43. Weekman, E.M.; Sudduth, T.L.; Caverly, C.N.; Kopper, T.J.; Phillips, O.W.; Powell, D.K.; Wilcock, D.M. Reduced efficacy of anti-A immunotherapy in a mouse model of amyloid deposition and vascular cognitive impairment comorbidity. J. Neurosci. 2016, 36, 9896-9907. PMid:27656027

    View Article      PubMed/NCBI     
  44. Nelson, A.R.; Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer's disease. Biochim. Biophys. Acta 2016, 1862, 887-900. PMid:26705676

    View Article      PubMed/NCBI     
  45. Kapasi, A.; Schneider, J.A. Vascular contributions to cognitive impairment, clinical Alzheimer's disease, and dementia in older persons. Biochim. Biophys. Acta 2016, 1862, 878-886. PMid:26769363

    View Article      PubMed/NCBI     
  46. McAleese, K.L.; Alafuzoff, I.; Charidimou, A.; De Reuck, J.; Grinberg, L.T.; Hainsworth, A.H.; Hortobagyi, T.; Ince, P.; Jellinger, K.; Gao, J.; et al. Post-mortem assessment in vascular dementia: Advances and aspirations. BMC Med. 2016, 14, 129. PMid:27600683

    View Article      PubMed/NCBI     
  47. Noh, Y.; Seo, S.W.; Jeon, S.; Lee, J.M.; Kim, J.S.; Lee, J.H.; Kim, J.H.; Kim, G.H.; Ye, B.S.; Cho, H.; et al. The role of cerebrovascular disease in amyloid deposition. J. Alzheimers Dis. 2016, 54, 1015-1026. PMid:27567803

    View Article      PubMed/NCBI     
  48. Hishikawa, N.; Fukui, Y.; Sato, K.; Kono, S.; Yamashita, T.; Ohta, T.; Deguchi, K.; Abe, K. Cognitive and affective functions in Alzheimer's disease patients with metabolic syndrome. Eur. J. Neurol. 2016, 23, 339-345. PMid:26493280

    View Article      PubMed/NCBI     
  49. Gutierrez, J.; Honig, L.; Elkind, M.S.; Mohr, J.P.; Goldman, J.; Dwork, A.J.; Morgello, S.; Marshall, R.S. Brain arterial aging and its relationship to Alzheimer dementia. Neurology 2016, 86, 1507-1515. PMid:26984942

    View Article      PubMed/NCBI     
  50. Nagata, K.; Yamazaki, T.; Takano, D.; Maeda, T.; Fujimaki, Y.; Nakase, T.; Sato, Y. Cerebral circulation in aging. Ageing Res. Rev. 2016, 30, 49-60. PMid:27484894

    View Article      PubMed/NCBI     
  51. Calabrese, V.; Giordano, J.; Signorile, A.; Ontario, M.L.; Castorina, S.; de Pasquale, C.; Eckert, G.; Calabrese, E.J. Major pathogenic mechanisms in vascular dementia: Roles of cellular stress response and hormesis in neuroprotection. J. Neurosci. Res. 2016, 94, 1588-1603. PMid:27662637

    View Article      PubMed/NCBI     
  52. Toth, P.; Tarantini, S.; Csiszar, A.; Ungvari, Z.I. Functional vascular contributions to cognitive impairment and dementia: Mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H1-H20. PMid:27793855

    View Article      PubMed/NCBI     
  53. Devraj, K.; Poznanovic, S.; Spahn, C.; Schwall, G.; Harter, P.N.; Mittelbronn, M.; Antoniello, K.; Paganetti, P.; Muhs, A.; Heilemann, M.; et al. BACE-1 is expressed in the blood-brain barrier endothelium and is upregulated in a murine model of Alzheimer's disease. J. Cereb. Blood Flow Metab. 2016, 36, 1281-1294. PMid:26661166

    View Article      PubMed/NCBI     
  54. Chao, A.C.; Lee, T.C.; Juo, S.H.; Yang, D.I. Hyperglycemia increases the production of amyloid -peptide leading to decreased endothelial tight junction. CNS Neurosci. Ther. 2016, 22, 291-297. PMid:26842741

    View Article      PubMed/NCBI     
  55. Khalil, R.B.; Khoury, E.; Koussa, S. Linking multiple pathogenic pathways in Alzheimer's disease.World J. Psychiatry 2016, 6, 208-214. PMid:27354962

    View Article      PubMed/NCBI     
  56. Festoff, B.W.; Sajja, R.K.; van Dreden, P.; Cucullo, L. HGMB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer's disease. J. Neuroinflamm. 2016, 13, 194. PMid:27553758

    View Article      PubMed/NCBI     
  57. Gangoda, S.V.; Butlin, M.; Gupta, V.; Chung, R.; Avolio, A. Pulsatile stretch alters expression and processing of amyloid precursor protein in human cerebral endothelial cells. J. Hypertens. 2016, 34, e24.

    View Article           
  58. Roberts, A.M.; Jagadapillai, R.; Vaishnav, R.A.; Friedland, R.P.; Drinovac, R.; Lin, X.; Gozal, E. Increased pulmonary arteriolar tone associated with lung oxidative stress and nitric oxide in a mouse model of Alzheimer's disease. Physiol. Rep. 2016, 4, e12953. PMid:27604401

    View Article      PubMed/NCBI     
  59. Shang, S.; Yang, Y.M.; Zhang, H.; Tian, L.; Jiang, J.S.; Dong, Y.B.; Zhang, K.; Li, B.; Zhao,W.D.; Fang,W.G.; et al. Intracerebral GM-CSF contributes to transendothelial monocyte migration in APP/PS1 Alzheimer's disease mice. J. Cereb. Blood Flow Metab. 2016, 36, 1987-1991. PMid:27444968

    View Article      PubMed/NCBI     
  60. Austin, S.A.; Katusic, Z.S. Loss of endothelial nitric oxide synthase promotes p25 generation and tau phosphorylation in amurinemodel of Alzheimer's disease. Circ. Res. 2016, 119, 1128-1134. PMid:27601478

    View Article      PubMed/NCBI     
  61. Katusic, Z.S.; Austin, S.A. Neurovascular protective function of endothelial nitric oxide. Circ. J. 2016, 80, 1499-1503. PMid:27238834

    View Article      PubMed/NCBI     
  62. Wang, L.; Du, Y.; Wang, K.; Xu, G.; Luo, S.; He, G. Chronic cerebral hypoperfusion induces memory deficits and facilitates A generation in C57BL/6J mice. Exp. Neurol. 2016, 283, 353-364. PMid:27421879

    View Article      PubMed/NCBI     
  63. Kyrtsos, C.R.; Baras, J.S. Modeling the role of the glymphatic pathway and cerebral blood vessel properties in Alzheimer's disease pathogenesis. PLoS ONE 2015, 10, e0139574. PMid:26448331

    View Article      PubMed/NCBI     
  64. Kalaria, R.N.; Akinyemi, R.; Ihara, M. Stroke injury, cognitive impairment and vascular dementia. Biochim. Biophys. Acta 2016, 1862, 915-925. PMid:26806700

    View Article      PubMed/NCBI     
  65. Khan, A.; Kalaria, R.N.; Corbett, A.; Ballard, C. Update on vascular dementia. J. Geriatr. Psychiatry Neurol. 2016, 29, 281-301. PMid:27502303

    View Article      PubMed/NCBI     
  66. Austin, S.A.; Santhanam, A.V.; d'Uscio, L.V.; Katusic, Z.S. Regional heterogeneity of cerebral microvessels and brain susceptibility to oxidative stress. PLoS ONE 2015, 10, e0144062. PMid:26629821

    View Article      PubMed/NCBI     
  67. Toda, N.; Okamura, T. Cigarette smoking impairs nitric oxide-mediated cerebral blood flow increase: Implications for Alzheimer's disease. J. Pharmacol. Sci. 2016, 131, 223-232. PMid:27530818

    View Article      PubMed/NCBI     
  68. Uiterwijk, R.; Huijts, M.; Staals, J.; Rouhl, R.P.; De Leeuw, P.W.; Kroon, A.A.; van Oostenbrugge, R.J. Endothelial activation is associated with cognitive performance in patients with hypertension. Am. J. Hypertens. 2016, 29, 464-469. PMid:26271106

    View Article      PubMed/NCBI     
  69. Kamat, P.K.; Kyles, P.; Kalani, A.; Tyagi, N. Hydrogen sulfide ameliorates homocysteine-induced Alzheimer's disease-like pathology, blood-brain barrier disruption, and synaptic disorder. Mol. Neurobiol. 2016, 53, 2451-2467. PMid:26019015

    View Article      PubMed/NCBI     
  70. Iadecola, C. Untangling neurons with endothelial nitric oxide. Circ. Res. 2016, 119, 1052-1054. PMid:27789581

    View Article      PubMed/NCBI     
  71. Wang, Y.J. Lessons from immunotherapy for Alzheimer's disease. Nat. Rev. Neurol. 2014, 10, 188-189. PMid:24638135

    View Article      PubMed/NCBI     
  72. Krstic, D.; Knuesel, I. Deciphering the mechanism underlying late-onset Alzheimer's disease. Nat. Rev. Neurol. 2013, 9, 25-34. PMid:23183882

    View Article      PubMed/NCBI     
  73. D'Arrigo, J. Stable Nanoemulsions: Self-Assembly in Nature and Nanomedicine; Elsevier: Amsterdam, The Netherlands, 2011; 415p, ISBN 978-0-444-53798-0.

  74. Barbarese, E.; Ho, S.Y.; D'Arrigo, J.S.; Simon, R.H. Internalization of microbubbles by tumor cells in vivo and in vitro. J. Neurooncol. 1995, 26, 25-34. PMid:8583242

    View Article      PubMed/NCBI     
  75. Camacho, J.; Moline, T.; Bonaterra-Pastra, A.; Ramon y Cajal, S.; Martinez-Saez, E.; Hernandez-Guillamon, M. Brain apoA-I, apoJ and apoE immunodetection in cerebral amyloid angiopathy. Front. Neurol. 2019, 10, 187. PMid:30918495

    View Article      PubMed/NCBI     
  76. D'Arrigo, J. Surfactant Mixtures, Stable Gas-in-Liquid Emulsions, and Methods for the Production of such Emulsions from Said Mixtures. U.S. Patent No. 4,684,479A , 4 August 1987.

  77. Garg, G.; Saraf, Sh.; Saraf, Sw. Cubosomes: An overview. Biol. Pharm. Bull. 2007, 30, 350-353. PMid:17268078

    View Article      PubMed/NCBI     
  78. Tanford, C. The Hydrophobic Effect: Formation of Micelles and Biological Membranes; Wiley: New York, NY, USA, 1973; 200p.

  79. Boyd, B.J.; Whittaker, D.V.; Khoo, S.M.; Davey, G. Lyotropic liquid crystalline phases formed from glycerate surfactants as sustained release drug delivery systems. Int. J. Pharm. 2006, 309, 218-226. PMid:16413980

    View Article      PubMed/NCBI     
  80. Pouton, C.W. Properties and uses of common formulation lipids, surfactants and cosurfactants. In Proceedings of the AAPSWorkshop, Effective Utilization of Lipid-Based Systems to Enhance the Delivery of Poorly Soluble Drugs: Physicochemical, Biopharmaceutical and Product Development Considerations, Bethesda, MD, USA, 5-6 March 2007; Constantinides, P.P., Porter, C.J.H., Eds.; AAPS: Arlington, VA, USA, 2007.

  81. Small, D.M. The behavior of biological lipids. Pure Appl. Chem. 1981, 53, 2095-2103.

    View Article           
  82. Kaasgaard, T.; Drummond, C.J. Ordered 2-D and 3-D nano-structured amphiphile self-assembly materials stable in excess solvent. Phys. Chem. Chem. Phys. 2006, 8, 4957-4975. PMid:17091149

    View Article      PubMed/NCBI     
  83. Shearman, G.C.; Khoo, B.J.; Motherwell, M.L.; Brakke, K.A.; Ces, O.; Conn, C.E.; Seddon, J.M.; Templer, R.H. Calculations of and evidence for chain packing stress in inverse lyotropic bicontinuous cubic phases. Langmuir 2007, 23, 7276-7285. PMid:17503862

    View Article      PubMed/NCBI     
  84. Rizwan, S.B.; Dong, Y.D.; Boyd, B.J.; Rades, T.; Hook, S. Characterization of bicontinuous cubic liquid crystalline systems of phytantriol and water using cryo field emission scanning electron microscopy (cryo FESEM). Micron 2007, 38, 478-485. PMid:17011783

    View Article      PubMed/NCBI     
  85. Yaghmur, A.; de Campo, L.; Sagalowicz, L.; Leser, M.E.; Glatter, O. Emulsified microemulsions and oil-containing liquid crystalline phases. Langmuir 2005, 21, 569-577. PMid:15641825

    View Article      PubMed/NCBI     
  86. Yaghmur, A.; de Campo, L.; Sagalowicz, L.; Leser, M.E.; Glatter, O. Control of the internal structure of MLO-based isasomes by the addition of diglycerol monooleate and soybean phosphatidylcholine. Langmuir 2006, 22, 9919-9927. PMid:17106981

    View Article      PubMed/NCBI     
  87. Gustafsson, J.; Ljusberg-Wahren, H.; Almgren, M.; Larsson, K. Submicron particles of reversed lipid phases in water stabilized by a nonionic amphiphilic polymer. Langmuir 1997, 13, 6964-6971. +

    View Article           
  88. De Campo, L.; Yaghmur, A.; Sagalowicz, L.; Leser, M.E.; Watzke, H.; Glatter, O. Reversible phase transitions in emulsified nanostructured lipid systems. Langmuir 2004, 20, 5254-5261. PMid:15986660

    View Article      PubMed/NCBI     
  89. Yaghmur, A.; de Campo, L.; Salentinig, S.; Sagalowicz, L.; Leser, M.E.; Glatter, O. Oil-loaded monolinolein-based particles with confined inverse discontinuous cubic structure (Fd3m). Langmuir 2006, 22, 517-521. PMid:16401095

    View Article      PubMed/NCBI     
  90. Larsson, K. Aqueous dispersions of cubic lipid-water phases. Curr. Opin. Colloid Interface Sci. 2000, 5, 64-69. 00040-6

    View Article           
  91. Luzzati, V. Biological significance of lipid polymorphism: The cubic phases. Curr. Opin. Struct. Biol. 1997, 7, 661-668. 80075-9

    View Article           
  92. Sagar, G.H.; Arunagirinathan, M.A.; Bellare, J.R. Self-assembled surfactant nano-structures important in drug-delivery: A review. Indian J. Exp. Biol. 2007, 45, 133-159.

  93. Anton, N.; Benoit, J.P.; Saulnier, P. Design and production of nanoparticles formulated from nano-emulsion templates: A review. J. Control. Release 2008, 128, 185-199. PMid:18374443

    View Article      PubMed/NCBI     
  94. Bansal, T.; Mustafa, G.; Khan, Z.I.; Ahmad, F.J.; Khar, R.K.; Talegaonkar, S. Solid self-nanoemulsifying delivery systems as a platform technology for formulation of poorly soluble drugs. Crit. Rev. Ther. Drug Carrier Syst. 2008, 25, 63-116. PMid:18540836

    View Article      PubMed/NCBI     
  95. Sadurni, N.; Solans, C.; Azemar, N.; Garcia-Celma, M.J. Studies on the formation of O/W nano-emulsions, by low-energy emulsification methods, suitable for pharmaceutical applications. Eur. J. Pharm. Sci. 2005, 26, 438-445. PMid:16153811

    View Article      PubMed/NCBI     
  96. Tresset, G. The multiple faces of self-assembled lipidic systems. PMC Biophys. 2009, 2, 3. PMid:19374753

    View Article      PubMed/NCBI     
  97. Hato, M.; Yamashita, J.; Shiono, M. Aqueous phase behavior of lipids with isoprenoid type hydrophobic chains. J. Phys. Chem. B 2009, 113, 10196-10209. PMid:19572621

    View Article      PubMed/NCBI     
  98. Barauskas, J.; Cervin, C.; Tiberg, F.; Johnsson, M. Structure of lyotropic self-assembled lipid nonlamellar liquid crystals and their nanoparticles in mixtures of phosphatidyl choline and -tocopherol (vitamin E). Phys. Chem. Chem. Phys. 2008, 10, 6483-6485. PMid:18979032

    View Article      PubMed/NCBI     
  99. Efrat, R.; Aserin, A.; Garti, N. On structural transitions in a discontinuous micellar cubic phase loaded with sodium diclofenac. J. Colloid Interface Sci. 2008, 321, 166-176. PMid:18279886

    View Article      PubMed/NCBI     
  100. Yaghmur, A.; Laggner, P.; Almgren, M.; Rappolt, M. Self-assembly in monoelaidin aqueous dispersions: Direct vesicles to cubosomes transition. PLoS ONE 2008, 3, e3747. PMid:19015726

    View Article      PubMed/NCBI     
  101. Yaghmur, A.; Glatter, O. Characterization and potential applications of nanostructured aqueous dispersions. Adv. Colloid Interface Sci. 2009, 147-148, 333-342. PMid:18804754

    View Article      PubMed/NCBI     
  102. Vandoolaeghe, P.; Rennie, A.R.; Campbell, R.A.; Nylander, T. Neutron reflectivity studies of the interaction of cubic phase nanoparticles with phospholipid bilayers of different coverage. Langmuir 2009, 25, 4009-4020. PMid:19714826

    View Article      PubMed/NCBI     
  103. Vandoolaeghe, P.; Barauskas, J.; Johnsson, M.; Tiberg, F.; Nylander, T. Interaction between lamellar (vesicles) and nonlamellar lipid liquid-crystalline nanoparticles as studied by time-resolved small-angle X-ray diffraction. Langmuir 2009, 25, 3999-4008. PMid:19714888

    View Article      PubMed/NCBI     
  104. Yaghmur, A.; Kriechbaum, M.; Amenitsch, H.; Steinhart, M.; Laggner, P.; Rappolt, M. Effects of pressure and temperature on the self-assembled fully hydrated nanostructures of monoolein-oil systems. Langmuir 2010, 26, 1177-1185. PMid:19681634

    View Article      PubMed/NCBI     
  105. Fong, W.K.; Hanley, T.; Boyd, B.J. Stimuli responsive liquid crystals provide "on-demand" drug delivery in vitro and in vivo. J. Control. Release 2009, 135, 218-226. PMid:19331865

    View Article      PubMed/NCBI     
  106. Amselem, S.; Friedman, D. Solid Fat Nanoemulsions. U.S. Patent No. 5,662,932A , 2 September 1997.

  107. Kuntsche, J.; Koch, M.H.J.; Drechsler, M.; Bunjes, H. Crystallization behavior of supercooled smectic cholesteryl myristate nanoparticles containing phospholipids as stabilizers. Colloids Surf. B Biointerfaces 2005, 44, 25-35. PMid:15990283

    View Article      PubMed/NCBI     
  108. Kuntsche, J.; Westesen, K.; Drechsler, M.; Koch, M.H.J.; Bunjes, H. Supercooled smectic nanoparticles: A potential novel carrier system for poorly water soluble drugs. Pharm. Res. 2004, 21, 1834-1843. PMid:15553230

    View Article      PubMed/NCBI     
  109. Nimmrich, V.; Eckert, A. Calcium channel blockers and dementia. Brit. J. Pharmacol. 2013, 169, 1203-1210. PMid:23638877

    View Article      PubMed/NCBI     
  110. Shirwany, N.A.; Payette, D.; Xie, J.; Guo, Q. The amyloid beta ion channel hypothesis of Alzheimer's disease. Neuropsychiatr. Dis. Treat. 2007, 3, 597-612.

  111. Di Scala, C.; Yahi, N.; Boutemeur, S.; Flores, A.; Rodriguez, L; Chahinian, H.; Fantini, J. Common molecular mechanism of amyloid pore formation by Alzheimer's β-amyloid peptide and α-synuclein. Sci. Rep. 2016, 6, 28781. PMid:27352802

    View Article      PubMed/NCBI     
  112. Demuro, A.; Smith, M.; Parker, I. Single-channel Ca2+ imaging implicates Aβ1-42 amyloid pores in Alzheimer's disease pathology. J. Cell Biol. 2011, 195, 515-524. PMid:22024165

    View Article      PubMed/NCBI     
  113. Serra-Batiste, M.; Ninot-Pedrosa, M.; Bayoumi, M.; Gairi, M.; Maglia, G.; Carulla, N. Aβ42 assembles into specific β-barrel pore-forming oligomers in membrane-mimicking environments. Proc. Natl. Acad. Sci. USA 2016, 113, 10866-10871. PMid:27621459 PMCid:PMC5047179

    View Article      PubMed/NCBI     
  114. Bode, D.C.; Baker, M.D.; Viles, J.H. Ion channel formation by amyloid-β42 oligomers but not amyloid-β40 in cellular membranes. J. Biol. Chem. 2017, 292, 1404-1413. PMid:27927987

    View Article      PubMed/NCBI     
  115. Di Scala, C.; Chahinian, H.; Yahi, N.; Garmy, N.; Fantini, J. Interaction of Alzheimer's β-amyloid peptides with cholesterol: Mechanistic insights into amyloid pore formation. Biochemistry 2014, 53, 4489-4502. PMid:25000142

    View Article      PubMed/NCBI     
  116. Ghosal, K.; Haag, M.; Verghese, P.B.; West, T.; Veenstra, T.; Braunstein, J.B.; Bateman, R.J.; Holtzman, D.M.; Landreth, G.E. Arandomized controlled study to evaluate the effect of bexarotene on amyloid-β and apolipoprotein E metabolism in healthy subjects. Alzheimers Dement. (NY) 2016, 2, 110-120. PMid:29067298

    View Article      PubMed/NCBI     
  117. Pierrot, N.; Lhommel, R.; Quenon, L.; Hanseeuw, B.; Dricot, L.; Sindic, C.; Maloteaux, J.M.; Octave, J.N.; Ivanoiu, A. Targretin [bexarotene] improves cognitive and biological markers in a patient with Alzheimer's disease. J. Alzheimer's Dis. 2016, 49, 271-276. PMid:26444777

    View Article      PubMed/NCBI     
  118. Mirza, Z.; Beg, M.A. Possible molecular interactions of bexarotene - a retinoid drug and Alzheimer's Aβ peptide: A docking study. Curr. Alzheimer Res. 2017, 14, 327-334.

  119. Huy, P.D.Q.; Thai, N.Q.; Bednarikova, Z.; Phuc, L.H.; Linh, H.Q.; Gazova, Z.; Li, M.S. Bexarotene does not clear amyloid beta plaques but delays fibril growth: Molecular mechanisms. ACS Chem. Neurosci. 2017, 8, 1960-1969. PMid:28689412

    View Article      PubMed/NCBI     
  120. Mariani, M.M.; Malm, T.; Lamb, R.; Jay, T.R.; Neilson, L.; Casali, B.; Medarametla, L.; Landreth, G.E. Neuronally-directed effects of RXR activation in a mouse model of Alzheimer's disease. Sci. Rep. 2017, 7, 42270. PMid:28205585

    View Article      PubMed/NCBI     
  121. Habchi, J.; Arosio, P.; Perni, M.; Costa, A.R.; Yagi-Utsumi, M.; Joshi, P.; Chia,S.; Cohen, S.I.A.; Muller, M.B.D.; Linse, S.; et al. An anticancer drug suppresses the primary nucleation reaction that initiates the production of the toxic Aβ42 aggregates linked with Alzheimer's disease. Sci. Adv. 2016, 2, e1501244. PMid:26933687

    View Article      PubMed/NCBI     
  122. Fantini, J.; Di Scala, C.; Yahi, N.; Troadec, J.D.; Sadelli, K.; Chahinian, H.; Garmy, N. Bexarotene blocks calcium-permeable ion channels formed by neurotoxic Alzheimer's β-amyloid peptides. ACS Chem. Neurosci. 2014, 5, 216-224. PMid:24383913

    View Article      PubMed/NCBI     
  123. Casali, B.T.; Reed-Geaghan, E.G.; Landreth, G.E. Nuclear receptor agonist-driven modification of inflammation and amyloid pathology enhances and sustains cognitive improvements in a mouse model of Alzheimer's disease. J. Neuroinflamm. 2018, 15, 43. PMid:29448961

    View Article      PubMed/NCBI     
  124. Tu, L.; Yang, X.L.; Zhang, Q.; Wang, Q; Tian, T.; Liu, D.; Qu, X.; Tian, J.Y. Bexarotene attenuates early brain injury via inhibiting microglia activation through PPARγ after experimental subarachnoid hemorrhage. Neurol. Res. 2018, doi:10.1080/01616412.2018.1463900 . PMid:29688151

    View Article      PubMed/NCBI     
  125. Dheer, Y.; Chitranshi, N.; Gupta, V.; Abbasi, M.; Mirzaei, M.; You, Y; Chung, R.; Graham, S.L.; Gupta, V. Bexarotene modulates retinoid-X-receptor expression and is protective against neurotoxic endoplasmic reticulum stress response and apoptotic pathway activation. Mol. Neurobiol. 2018, doi:10.1007/s12035-018-1041-9 . PMid:29637440

    View Article      PubMed/NCBI     
  126. Kamp, F.; Scheidt, H.A.; Winkler, E.; Basset, G.; Heinel, H.; Hutchison, J.M.; LaPointe, L.M.; Sanders, C.R.; Steiner, H.; Huster, D. Bexarotene binds to the amyloid precursor protein transmembrane domain, alters its α-helical conformation, and inhibits γ-secretase nonselectivity in liposomes. ACS Chem. Neurosci. 2018, doi:10.1021acschemneuro.8b00068 .

  127. Serra-Batiste, M.; Tolchard, J.; Giusti, F.; Zoonens, M.; Carulla, N. Stabilization of a membrane-associated amyloid-β oligomer for its validation in Alzheimer's disease. Front. Mol. Biosci. 2018, 5, 38. PMid:29725595

    View Article      PubMed/NCBI     
  128. Xiang, N.; Lyu, Y.; Zhu, X.; Narsimhan, G. Investigation of the interaction of amyloid-β peptide (11-42) oligomers with a 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) membrane using molecular dynamics simulation. Phys. Chem. Chem.Phys. 2018, 20, 6817-6829. PMid:29299557

    View Article      PubMed/NCBI     
  129. Habchi, J.; Chia, S.; Galvagnion, C.; Michaels, T.C.T.; Bellaiche, M.M.J.; Ruggeri, F.S.; Sanguanini, M.; Idini, I.; Kumita, J.R.; Sparr, E.; et al. Cholesterol catalyses Aβ42 aggregation through a heterogeneous nucleation pathway in the presence of lipid membranes. Nat. Chem. 2018, 10, 673-683. PMid:29736006

    View Article      PubMed/NCBI     
  130. Bowman, G.L.; Dayon, L.; Kirkland, R.; Wojcik, J.; Peyratout, G.; Severin, I.C.; Henry, H.; Oikonomidi, A.; Migliavacca, E.; Bacher, M.; Popp, J. Blood-brain barrier breakdown, neuroinflammation, and cognitive decline in older adults. Alzheimer Dementia (online) 2018, doi:10.1016/j.jalz.2018.06.2857 . PMid:30120040

    View Article      PubMed/NCBI     
  131. Wang, H.; Golob, E.J.; Su, M.Y. Vascular volume and blood-brain barrier permeability measured by dynamic contrast enhanced MRI in hippocampus and cerebellum of patients with MCI and normal controls. J. Magn. Reson. Imaging 2006, 24, PMid:16878309

    View Article      PubMed/NCBI     
  132. Montagne, A.; Barnes, S.R.; Sweeney, M.D.; Halliday, M.R.; Sagare, A.P.; Zhao, Z.; Toga, A.W.; Jacobs, R.E.; Liu, C.Y.; Amezcua, L.; et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015, 85, 296-302. PMid:25611508

    View Article      PubMed/NCBI     

Journal Recent Articles