Alzheimers Disease & Dementia

ISSN: 2578-6490

RESEARCH ARTICLE | VOLUME 5 | ISSUE 1 | DOI: 10.36959/734/381 OPEN ACCESS

Drug Nanotargeting for Treating Vascular Dementia and Alzheimer's Disease

Joseph S. D'Arrigo, PhD*

  • Joseph S. D'Arrigo 1*
  • Cavitation-Control Technology Inc., Farmington, USA

D'Arrigo J (2021) Drug Nanotargeting for Treating Vascular Dementia and Alzheimer's Disease. Alzheimers Dis Dement 5(1):113-118.

Accepted: February 01, 2021 | Published Online: February 03, 2021

Drug Nanotargeting for Treating Vascular Dementia and Alzheimer's Disease

Abstract


Microvascular endothelial dysfunction precedes, often by decades, the cognitive decline associated with Alzheimer's disease. Hence, preservation of a healthy cerebrovascular endothelium can be an important therapeutic target. By incorporating appropriate drug(s) into biobased (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic which targets certain cell-surface scavenger receptors, mainly class B type I (i.e., SR-BI), and crosses the blood-brain barrier. Such colloidal-nanocarrier targeting allows for various Alzheimer's-related cell types to be simultaneously searched in a holistic integrative approach, in vivo, for localized drug treatment. Using various biobased 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, monoglyceride-based lyotropic liquid-crystalline phases are relatively unique owing to their rich polymorphism in water and potential application as drug nanocarriers. This (colloidal-nanocarrier) in vivo targeting advantage may be particularly important when delivering pleiotropic natural substances (e.g., an isoflavone) or for repurposing an FDA-approved drug.

Keywords


Alzheimer's disease, Blood-brain barrier, Cognitive impairment, Drug targeting, Lipid cubic phases, Nanoemulsion, Scavenger receptors, SR-BI, Vascular dementia

Introduction


Vascular brain lesions are very common in people over 70-years-old, and recent reviews [1,2] provide much evidence that a large proportion of dementia cases may be attributable to cerebrovascular disease [3,4]. Accordingly, vascular cognitive impairment and dementia (VCID) is the second leading cause of dementia behind Alzheimer's disease, and is a frequent co-morbidity in the Alzheimer's patient [5,6]. Furthermore, growing data from brain imaging studies and various animal models suggest that cerebrovascular dysfunction may well preceed cognitive impairment and the onset of neurodegenerative changes in Alzheimer's disease [2,4].

Endothelial dysfunction and targeted nanotherapy for early dementia

Small-vessel disease is commonly found in patients who have other brain pathologies, such as plaques and tangles associated with neurodegenerative disease. The vascular changes associated with small-vessel disease include alterations in density and morphology of cerebral microvasculature, and a blood-brain barrier (BBB) breakdown with leakage of blood-borne molecules [4]. It is no surprise, therefore, that multiple epidemiological studies have shown a marked overlap among risk factors for small-vessel cerebrovascular disease and late-onset Alzheimer's disease.

It has been reported repeatedly that endothelial modulation and repair is feasible by pharmacological targeting [1,2,7-13] via SR-BI receptors [13] As the detailed review by Mahringer, et al. [14] points out, the BBB is equipped with several endocytic receptors at the luminal surface (i.e., the capillary endothelial membrane), including SR-BI. Recently, Fung, et al. [15] specifically found that SR-BI mediates the uptake and transcytosis of high-density lipoproteins (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 [16-18]. Documented similarities in lipid composition - among HDL and the biomimetic (nanoemulsion) nanocarrier particles - can partially simulate or mimic the known heterogeneity (i.e., subpopulations or subspecies) of HDL particles [1,2].

This targeted-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 [1,19-29] and an apparent endothelium dysfunction [2,17,18,25,30-36] in both Alzheimer's disease and its major risk factors [1,2,29-41]. By incorporating drug molecules into the LCM/ND lipid nanoemulsion type (yielding particle sizes mostly < 0.1 μm in diameter), known to be a successful drug carrier, [42,43] one is likely to obtain a multitasking combination therapeutic capable of targeting cell-surface SR-BI. This (intravenous) combination therapeutic would make it possible for various cell types, all potentially implicated in Alzheimer's disease, [1,2] to be simultaneously sought out and better reached for localized drug treatment of brain tissue in vivo [42,43].

LCM/ND nanoemulsion type and targeting via lipid cubic phases

Importantly, monoglyceride is the largest single-lipid fraction (by wt. %) of the powdered solid lipid surfactants used to produce the (Filmix®) LCM/ND nanoemulsions [42] As a group, monoglycerides exhibit different phase behaviors when they are exposed to water [44,45]. 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 [42]. The (lyotropic or solvent-induced) cubic liquid-crystalline phases may be classified into two distinct classes: Bicontinuous cubic phases [46] and micellar or discontinuous (e.g., type Fd3m) cubic phases [47].

A noteworthy lipid cubic phase of the latter category is based upon packings of discrete inverse micellar aggregates and is formed by a variety of lipid systems [47]. Seddon, et al. [48] point out that the most frequently observed such (inverse micellar cubic) structure is a cubic phase of crystallographic space group Fd3m. Luzzati and coworkers have reported that this Fd3m cubic phase evidently requires a heterogeneous mixture of polar lipids [49,50]. 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 [51,52]) specifically include cholesterol and three categories of (saturated) glycerides, that is, tri-, di-, and monoglycerides [51,52]. In view of the advantageous attributes of monoglycerides (recounted above), 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 [42].

In this particular targeted-delivery approach, the self-assembled "lipid particle" structure itself (upon intravenousinjection of the LCM/ND nanoemulsion) is apparently successfully utilized as the "active" targeting ligand -which is directed via (adsorption of) plasma lipoproteins (including notably apoA-I) 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 [42].

Amyloid-β ion channel hypothesis of Alzheimer's disease

As explained in many reviews [53,54] 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+ [53,54].

Historical support for the above amyloid-β ion channel hypothesis, or so-called "calcium hypothesis", has also been observed at the clinical level [55]. 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 [56]. (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.) Hence, neurotoxicity is associated with soluble aggregates (i.e., oligomers) of Aβ rather than with the plaques themselves [56].

As summarized by Di Scala, et al., [55] 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 [57] (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 [57] Very recently, a different research group reported very similar findings [58]. 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, these authors' own detailed experimental data [58] 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 [58].

Brain injury, edaravone, and Alzheimer's disease

Besides the above considerations about amyloid pore formation, another important pathophysiological overlap is described, in the literature, existing between brain injury and Alzheimer's-disease brain. Wang, et al. [59] have pointed out that non-neuronal brain cells, especially astrocytes (the predominant cell type in the human brain), may exert an active role in the pathogenesis of traumatic brain injury (TBI). Activated astrocytes may contribute to increased oxidative stress and neuroinflammation following neurotrauma. Interestingly, the drug Edaravone has been used successfully, in past research, due to its neuroprotective and antioxidative effects on the brain after TBI. Wang, et al. [59] extended this research and found that, after intravenous administration (in rats), Edaravone treatment significantly decreased hippocampal neuron loss, reduced oxidative stress, and decreased neuronal programmed cell death as compared with control treatment. The protective effects of Edaravone treatment were also related to the pathology of TBI on non-neuronal cells, as Edaravone decreased both astrocyte and microglia activation following TBI. These authors conclude that the likely mechanism of Edaravone's neuroprotective effect in the rat model of TBI is via inhibiting oxidative stress, leading to a decreased inflammatory response and decreased glial activation, and thereby reducing neuronal death and improving neurological function. [59] Similarly, Itoh, et al. [60] have reported that intravenous Edaravone administration (in rats), following TBI, inhibited free radical-induced neuronal degeneration and apoptotic cell death around the damaged area. Hence, Edaravone treatment improved cerebral dysfunction following TBI, suggesting its potential as an effective clinical therapy [60].

In view of the above description of TBI, the effects of the drug Edaravone, and the pathophysiological overlap of TBI with many characteristics of Alzheimer's disease brain (cf. above), it is logical and consistent that Jiao, et al. [61] have recently reported that Edaravone can also ameliorate Alzheimer's disease-type pathologies and cognitive deficits of a mouse model of Alzheimer's disease. Specifically, besides reducing amyloid deposition and tau hyperphosphorylation, Edaravone was found to alleviate oxidative stress and, hence, attenuates the downstream pathologies including glial activation, neuroinflammation, neuronal loss, and synaptic dysfunction, and rescues the memory deficits of the mice [61]. (Note that Edaravone is a small-molecule drug, which is known to function as a free-radical scavenger, it currently is being used clinically in Japan to treat (acute ischemic) stroke patients. [59,61]) Jiao, et al. [61] further state that their above findings suggest that Edaravone is a promising drug candidate for Alzheimer's disease by targeting multiple key pathways of the disease pathogenesis. This recommendation by Jiao, et al. of Edaravone (for treating Alzheimer's disease) fits well with the initial drug candidates suggested elsewhere, [1] based on low-molecular-weight and sufficient lipophilicity, for incorporation into the LCM/ND lipid nanoemulsion (as again proposed here) to treat Alzheimer's disease. Since the Jiao, et al. recommendation is based in part on knowledge of failed clinical trials indicating that a single target or pathway does not work on this complex disease, [61] these investigators are understandably encouraged by a drug like Edaravone which targets multiple pathways of Alzheimer's disease pathogenesis.

While the risk factors for dementia trigger widespread inflammation and oxidative stress (e.g., [62,63]), it is also true that these two processes can result in more biological effects than enhanced calcium load in brain tissue and neurodegeneration (cf. [64]). In fact, oxidative stress and inflammation each involve pathophysiological cascades associatedwith a wide range of pathologies and especially aging. However, these two processes/cascades are not always associated with biological damage. (For example, oxidative stress constitutes an important mechanism in many physiological processes, such as adaptations to physical exercise and cell signaling.) Yet, when oxidative stress and/or inflammation are dysregulated, their action is harmful [64,65]. (In this situation, one corresponding example [of many] occurs in Alzheimer's disease, where growing evidence links the "reactive oxygen species" (ROS)-mediated damages with molecular targets including mitochondrial dynamics/function, autophagic pathways, and proteostasis balance. [66]) Accordingly, Khalil, et al. [67] found that Alzheimer's disease impaired the interaction of HDL (and ApoA-I) with the SR-BI receptor, and their experimental results indicated that such patients had higher levels of oxidative stress [67,68]. The authors concluded that their clinical study provides evidence for the first time that the functionality of HDL is impaired in Alzheimer's disease, and that this alteration may be caused by Alzheimer's disease-associated oxidative stress and inflammation [67,68]. This conclusion is consistent with earlier work where SR-BI was identified on astrocytes and vascular smooth muscle cells in Alzheimer's disease brain, and has been demonstrated to mediate the adhesion of microglia to aggregated Aβ (cf. [68]). Moreover, these authors further report that SR-BI mediates perivascular macophage response, and regulates Aβ-related pathology and cerebral amyloid angiopathy, in an Alzheimer's-disease mouse model [68].

Concluding Remarks


Microvascular endothelial dysfunction precedes, often by decades, the cognitive decline associated with Alzheimer's disease. The cardiovascular risk factors for this disease trigger widespread inflammation and oxidative stress, both of which can lead to BBB disruption. Past studies (e.g., [69,70]) have shown that low-grade inflammation and endothelial dysfunction contribute to reduced information processing speed and executive functioning in an older population. These interacting processes involve pathophysiological cascades which lead to neuronal (intracellular) Ca2+ increase, neurodegeneration, gradual cognitive/memory decline, and eventually (late-onset) dementia. By incorporating 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 (i.e., SR-BI), and crosses the BBB. Such biomedical application ofcolloidal drug-nanocarriers can potentially be extended to thetreatment of complex medical disorders like vascular dementia and (late-onset) Alzheimer's disease [69,70]. Recent reviews (e.g., [66,71,72]) of human Alzheimer's-disease studies have noted a significant elevation in inflammatory mediators in the cerebral microcirculation, crucially, inflammation has a key role in linking several types of vascular and neuronal damage (in Alzheimer's-disease brain) to cardiovascular risk factors, such as arterial stiffness and hypertension [66]. Lastly, it has been reconfirmed in the current literature that receptor-mediated endocytosis/transcytosis via lipoprotein receptors, particularly scavenger receptors including SR-BI, remains a major route for drug delivery across the blood-brain barrier, namely, recently published work has demonstrated that nanocomplexes can be readily transported into brain capillary endothelial cells (bovine and porcine) via SR-BI receptor-mediated endocytosis [7] (see also [73-75]). Accordingly, endothelial modulation and repair become feasible by pharmacological targeting [8-12,76-79] via SR-BI receptors (cf. [13]). The proposed multitasking combination therapeutic appears likely to display greater efficacy at different stages of Alzheimer's disease [41]. Moreover, the effects of the various cell types targeted (via SR-BI) may be additive, multiplicative, or otherwise synergistic.

Acknowledgements


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

Conflicts of Interest


Authors declare no conflict of interest.J.S.D. is employed at Cav-Con Inc.

References


  1. D'Arrigo JS (2017) Alzheimer's disease, brain injury, and CNS nanotherapy in humans: Sonoporation augmenting drug targeting. Med Sci 5: 29.
  2. D'Arrigo JS (2018) Nanotherapy for Alzheimer's disease and vascular dementia: targeting senile endothelium. Adv Colloid Interface Sci 251: 44-54.
  3. Tariq S, d'Esterre CD, Sajobi TT, et al. (2018) A longitudinal magnetic resonance imaging study of neurodegenerative and small vessel disease, and clinical cognitive trajectories in non-demented patients with transient ischemic attack: the PREVENT study. BMC Geriatrics 18: 163.
  4. Stefanova NA, Maksimova KY, Rudnitskaya EA, et al. (2018) Association of cerebrovascular dysfunction with the development of Alzheimer's disease-like pathology in OXYS rats. BMC Genomics 19: 75.
  5. Kalaria RN (2016) Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer's disease. Acta Neuropathol 131: 659-685.
  6. Duncombe J, Kitamura A, Hase Y, et al. (2017) Chronic cerebral hypoperfusion: a key mechanism leading to vascular cognitive impairment and dementia Closing the translational gap between rodent models and human vascular cognitive impairment and dementia. Clin Sci (Lond) 131: 2451-2468.
  7. Srimanee A, Regberg J, Hallbrink M, et al. (2016) Role of scavenger receptors in peptide-based delivery of plasmid DNA across a blood-brain barrier model. Int J Pharm 500: 128-135.
  8. Di Marco LY, Venneri A, Farkas E, et al. (2015) Vascular dysfunction in the pathogenesis of Alzheimer's disease - A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis 82: 593-606.
  9. Carradori D, Gaudin A, Brambilla D, et al. (2016) Application of nanomedicine to the CNS diseases. Int Rev Neurobiol 130: 73-113.
  10. Zenaro E, Piacentino G, Constantin G (2016) The blood - brain barrier in Alzheimer's disease. Neurobiol Dis 107: 41-56.
  11. Qosa H, Mohamed A, Al Rihani SB, et al. (2016) High-throughput screening for identification of blood–brain barrier integrity enhancers: a drug repurposing opportunity to rectify vascular amyloid toxicity. J Alzheimers Dis 53: 1499-1516.
  12. Koizumi K, Wang G, Park L (2016) Endothelial dysfunction and amyloid-induced neurovascular alterations. Cell Mol Neurobiol 36: 155-165.
  13. Goldwaser EL, Acharya NK, Sarkar A, et al. (2016) Breakdown of the cerebrovasculature and blood-brain barrier: a mechanistic link between diabetes mellitus and Alzheimer's disease. J Alzheimers Dis 54: 445-456.
  14. Mahringer A, Reichel V, Ott M, et al. (2012) Overcoming the blood brain barrier: the challenge of brain drug targeting. J Nanoneurosci 2: 5-19.
  15. Fung KY, Wang C, Nyegaard S, et al. (2017) SR-BI mediated transcytosis of HDL in brain microvascular endothelial cells is independent of caveolin, clathrin, and PDZK1. Front Physiol 8: 841.
  16. Robert J, Button EB, Stukas S, et al. (2017) High-density lipoproteins suppress Aß-induced PBMC adhesion to human endothelial cells in bioengineered vessels and in monoculture. Mol Neurodegener 12: 60.
  17. Robert J, Stukas S, Button E, et al. (2016) Reconstituted high-density lipoproteins acutely reduce soluble brain Aß levels in symptomatic APP/PS1 mice. Biochim Biophys Acta 1862: 1027-1036.
  18. Hottman DA, Chernick D, Cheng S, et al. (2014) HDL and cognition in neurodegenerative disorders. Neurobiol Dis 72: 22-36.
  19. Weekman EM, Sudduth TL, Caverly CN, et al. (2016) Reduced efficacy of anti-A immunotherapy in a mouse model of amyloid deposition and vascular cognitive impairment comorbidity. J Neurosci 36: 9896-9907.
  20. Nelson AR, Sweeney MD, Sagare AP, et al. (2016) Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer's disease. Biochim Biophys Acta 1862: 887-900.
  21. Kapasi A, Schneider JA (2016) Vascular contributions to cognitive impairment, clinical Alzheimer's disease, and dementia in older persons. Biochim Biophys Acta 1862: 878-886.
  22. McAleese KL, Alafuzoff I, Charidimou A, et al. (2016) Post-mortem assessment in vascular dementia: Advances and aspirations. BMC Med 14: 129.
  23. Noh Y, Seo SW, Jeon S, et al. (2016) The role of cerebrovascular disease in amyloid deposition. J Alzheimers Dis 54: 1015-1026.
  24. Hishikawa N, Fukui Y, Sato K, et al. (2016) Cognitive and affective functions in Alzheimer's disease patients with metabolic syndrome. Eur J Neurol 23: 339-345.
  25. Gutierrez J, Honig L, Elkind MS, et al (2016) Brain arterial aging and its relationship to Alzheimer dementia. Neurology 86: 1507-1515.
  26. Nagata K, Yamazaki T, Takano D, et al. (2016) Cerebral circulation in aging. Ageing Res Rev 30: 49-60.
  27. Calabrese V, Giordano J, Signorile A, et al. (2016) Major pathogenic mechanisms in vascular dementia: roles of cellular stress response and hormesis in neuroprotection. J Neurosci Res 94: 1588-1603.
  28. Toth P, Tarantini S, Csiszar A, et al. (2017) 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 312: H1-H20.
  29. Devraj K, Poznanovic S, Spahn C, et al. (2016) 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 36: 1281-1294.
  30. Chao AC, Lee TC, Juo SH, et al. (2016) Hyperglycemia increases the production of amyloid-peptide leading to decreased endothelial tight junction. CNS Neurosci Ther 22: 291-297.
  31. Khalil RB, Khoury E, Koussa S (2016) Linking multiple pathogenic pathways in Alzheimer's disease. World J Psychiatry 6: 208-214.
  32. Festoff BW, Sajja RK, van Dreden P, et al. (2016) HGMB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer's disease. J Neuroinflamm 13: 194.
  33. Gangoda SV, Butlin M, Gupta V, et al. (2016) Pulsatile stretch alters expression and processing of amyloid precursor protein in human cerebral endothelial cells. J Hypertens 34: e24.
  34. Roberts AM, Jagadapillai R, Vaishnav RA, et al. (2016) Increased pulmonary arteriolar tone associated with lung oxidative stress and nitric oxide in a mouse model of Alzheimer's disease. Physiol Rep 4: e12953.
  35. Kyrtsos CR, Baras JS (2015) Modeling the role of the glymphatic pathway and cerebral blood vessel properties in Alzheimer's disease pathogenesis. PLoS One 10: e0139574.
  36. Kalaria RN, Akinyemi R, Ihara M (2016) Stroke injury, cognitive impairment and vascular dementia. Biochim Biophys Acta 1862: 915-925.
  37. Khan A, Kalaria RN, Corbett A, et al. (2016) Update on vascular dementia. J Geriatr Psychiatry Neurol 29: 281-301.
  38. Toda N, Okamura T (2016) Cigarette smoking impairs nitric oxide-mediated cerebral blood flow increase: implications for Alzheimer's disease. J Pharmacol Sci 131: 223-232.
  39. Uiterwijk R, Huijts M, Staals J, et al. (2016) Endothelial activation is associated with cognitive performance in patients with hypertension. Am J Hypertens 29: 464-469.
  40. Wang YJ (2014) Lessons from immunotherapy for Alzheimer's disease. Nat Rev Neurol 10: 188-189.
  41. Krstic D, Knuesel I (2013) Deciphering the mechanism underlying late-onset Alzheimer's disease. Nat Rev Neurol 9: 25-34.
  42. D'Arrigo JS (2011) Stable Nanoemulsions: Self-Assembly in Nature and Nanomedicine. Amsterdam: Elsevier, 415.
  43. Barbarese E, Ho SY, D'Arrigo JS, et al. (1995) Internalization of microbubbles by tumor cells in vivo and in vitro. J Neurooncol 26: 25-34.
  44. Garg G, Saraf SH, Saraf SW (2007) Cubosomes: an overview. Biol Pharm Bull 30: 350-353.
  45. (2007) Pouton CW Properties and uses of common formulation lipids, surfactants and cosurfactants. In: PP Constantinides, CJH Porter, Effective Utilization of Lipid-Based Systems to Enhance the Delivery of Poorly Soluble Drugs: Physicochemical, Biopharmaceutical and Product Development Considerations. Proceedings of the AAPSWorkshop, Arlington, VA, USA.
  46. Kaasgaard T, Drummond CJ (2006) Ordered 2-D and 3-D nano-structured amphiphile self-assembly materials stable in excess solvent. Phys Chem Chem Phys 8: 4957-4975.
  47. Small DM (1981) The behavior of biological lipids Pure Appl Chem 53: 2095-2103.
  48. Seddon JM, Robins J, Gulik-Krzywicki T, et al. (2000) Inverse micellar phases of phospholipids and glycolipids. Phys Chem Chem Phys 2: 4485-4493.
  49. Luzzati V, Vargas R, Mariani P, et al. (1993) Cubic phases of lipid-containing systems: elements of a theory and biological connotations. J Mol Biol 229: 540-551.
  50. Luzzati V, Vargas R, Gulik A, et al. (1992) Lipid polymorphism: a correction The structure of the cubic phase of extinction symbol Fd -consists of two types of disjointed reverse micelles embedded in a three-dimensional hydrocarbon matrix. Biochemistry 31: 279-285.
  51. D'Arrigo JS (1987) Surfactant mixtures, stable gas-in-liquid emulsions, and methods for the production of such emulsions from said mixtures.
  52. D'Arrigo JS (1993) Method for the production of medical-grade lipid-coated microbubbles, paramagnetic labeling of such microbubbles and therapeutic uses of microbubbles.
  53. Nimmrich V, Eckert A (2013) Calcium channel blockers and dementia. Brit J Pharmacol 169: 1203-1210.
  54. Shirwany NA, Payette D, Xie J, et al. (2007) The amyloid beta ion channel hypothesis of Alzheimer's disease. Neuropsychiatr Dis Treat 3: 597-612.
  55. Di Scala C, Yahi N, Boutemeur S, et al. (2016) Common molecular mechanism of amyloid pore formation by Alzheimer's ß-amyloid peptide and a-synuclein. Sci Rep 6: 28781.
  56. Demuro A, Smith M, Parker I (2011) Single-channel Ca2+ imaging implicates Aß1-42 amyloid pores in Alzheimer's disease pathology. J Cell Biol 195: 515-524.
  57. Serra-Batiste M, Ninot-Pedrosa M, Bayoumi M, et al. (2016) Aß42 assembles into specific ß-barrel pore-forming oligomers in membrane-mimicking environments. Proc Natl Acad Sci USA 113: 10866-10871.
  58. Bode DC, Baker MD, Viles JH (2017) Ion channel formation by amyloid-ß42 oligomers but not amyloid-ß40 in cellular membranes. J Biol Chem 292: 1404-1413.
  59. Wang GH, Jiang ZL, Li YC, et al. (2011) Free-radicle scavenger Edaravone treatment confers neuroprotection against traumatic brain injury in rats. J Neurotrauma 28: 2123-2134.
  60. Itoh T, Satou T, Nishida S, et al. (2010) Edaravone protects against apoptotic neuronal cell death and improves cerebral function after traumatic brain injury in rats. Neurochem Res 35: 348-355.
  61. Jiao SS, Yao XQ, Liu YH, et al. (2015) Edaravone alleviates Alzheimer's disease-type pathologies and cognitive deficits. Proc Natl Acad Sci USA 112: 5225-5230.
  62. Daulatzai MA (2016) Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer's disease. J Neurosci Res 95: 943-972.
  63. D'Arrigo JS (2020) Biomaterial to improve drug delivery in Alzheimer's disease: Linking major pathogenic pathways. OBM Geriatrics 4: 10.
  64. Gambini J (2020) Oxidative stress and inflammation: From mechanisms to therapeutic approaches Biomedicines.
  65. D'Arrigo JS (2020) Biomimetic nanocarrier targeting drug(s) to upstream-receptor mechanisms in dementia: Focusing on linking pathogenic cascades. Biomimetics 5: 11.
  66. Tangestani Fard M, Stough C (2019) A review and hypothesized model of the mechanisms that underpin the relationship between inflammation and cognition in the elderly. Front Aging Neurosci 11: 56.
  67. Khalil A, Berrougui H, Pawelec G, et al. (2012) Impairment of the ABCA1 and SR-BI-mediated cholesterol efflux pathways and HDL anti-inflammatory activity in Alzheimer's disease. Mech Ageing Dev 133: 20-29.
  68. Thanopoulou K, Fragkouli A, Stylianopoulou F, et al. (2010) Scavenger receptor class B type I (SR-BI) regulates perivascular macrophages and modifies amyloid pathology in an Alzheimer mouse model. Proc Natl Acad Sci 107: 20816-20821.
  69. Heringa SM, van den Ber E, Reijmer YD, et al. (2014) Markers of low-grade inflammation and endothelial dysfunction are related to reduced information processing speed and executive functioning in an older population -the Hoorn Study. Psychoneuroendocrinology 40: 108-118.
  70. D'Arrigo JS (2020) Nanotargeting dementia etiology: Aiming drug nanocarriers toward receptors for vascular endothelium, serum amyloid A, inflammasomes, and oxidative stress. Nano Prog 2: 25-30.
  71. Lenart N, Brough D, Denes A (2016) Inflammasomes link vascular disease with neuroinflammation and brain disorders. J Cereb Blood Flow Metab 36: 1668-1685.
  72. Sierksma A, Lu A, Mancuso R, et al. (2020) Novel Alzheimer risk genes determine the microglia response to amyloid-ß but not to TAU pathology. EMBO Mol Med 12: e10606.
  73. Lajoie JM, Shusta EV (2015) Targeting receptor-mediated transport for delivery of biologics across the blood-brain barrier .Annu Rev Pharmacol Toxicol 55: 613-631.
  74. Almer G, Mangge H, Zimmer A, et al. (2015) Lipoprotein-related and apolipoprotein-mediated delivery systems for drug targeting and imaging. Curr Med Chem 22: 3631-3651.
  75. Preston JE, Abbott J, Begley DJ (2014) Transcytosis of macromolecules at the blood-brain barrier. Adv Pharmacol 71: 147-163.
  76. Salmina AB, Inzhutova AI, Malinovskaya NA, et al. (2010) Endothelial dysfunction and repair in Alzheimer-type neurodegeneration: Neuronal and glial control. J Alzheimers Dis 22: 17-36.
  77. Tong XK, Hamel E (2015) Simvastatin restored vascular reactivity, endothelial function and reduced string vessel pathology in a mouse model of cerebrovascular disease. J Cereb Blood Flow Metab 35: 512-520.
  78. Koster KP, Thomas R, Morris AW, et al. (2016) Epidermal growth factor prevents oligomeric amyloid-induced angiogenesis deficits in vitro. J Cereb Blood Flow Metab 36: 1865-1871.
  79. Hostenbach S, D'haeseleer M, Kooijman R, et al. (2016) The pathophysiological role of astrocytic endothelin-1. Prog Neurobiol 144: 88-102.

Abstract


Microvascular endothelial dysfunction precedes, often by decades, the cognitive decline associated with Alzheimer's disease. Hence, preservation of a healthy cerebrovascular endothelium can be an important therapeutic target. By incorporating appropriate drug(s) into biobased (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic which targets certain cell-surface scavenger receptors, mainly class B type I (i.e., SR-BI), and crosses the blood-brain barrier. Such colloidal-nanocarrier targeting allows for various Alzheimer's-related cell types to be simultaneously searched in a holistic integrative approach, in vivo, for localized drug treatment. Using various biobased 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, monoglyceride-based lyotropic liquid-crystalline phases are relatively unique owing to their rich polymorphism in water and potential application as drug nanocarriers. This (colloidal-nanocarrier) in vivo targeting advantage may be particularly important when delivering pleiotropic natural substances (e.g., an isoflavone) or for repurposing an FDA-approved drug.

References

  1. D'Arrigo JS (2017) Alzheimer's disease, brain injury, and CNS nanotherapy in humans: Sonoporation augmenting drug targeting. Med Sci 5: 29.
  2. D'Arrigo JS (2018) Nanotherapy for Alzheimer's disease and vascular dementia: targeting senile endothelium. Adv Colloid Interface Sci 251: 44-54.
  3. Tariq S, d'Esterre CD, Sajobi TT, et al. (2018) A longitudinal magnetic resonance imaging study of neurodegenerative and small vessel disease, and clinical cognitive trajectories in non-demented patients with transient ischemic attack: the PREVENT study. BMC Geriatrics 18: 163.
  4. Stefanova NA, Maksimova KY, Rudnitskaya EA, et al. (2018) Association of cerebrovascular dysfunction with the development of Alzheimer's disease-like pathology in OXYS rats. BMC Genomics 19: 75.
  5. Kalaria RN (2016) Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer's disease. Acta Neuropathol 131: 659-685.
  6. Duncombe J, Kitamura A, Hase Y, et al. (2017) Chronic cerebral hypoperfusion: a key mechanism leading to vascular cognitive impairment and dementia Closing the translational gap between rodent models and human vascular cognitive impairment and dementia. Clin Sci (Lond) 131: 2451-2468.
  7. Srimanee A, Regberg J, Hallbrink M, et al. (2016) Role of scavenger receptors in peptide-based delivery of plasmid DNA across a blood-brain barrier model. Int J Pharm 500: 128-135.
  8. Di Marco LY, Venneri A, Farkas E, et al. (2015) Vascular dysfunction in the pathogenesis of Alzheimer's disease - A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis 82: 593-606.
  9. Carradori D, Gaudin A, Brambilla D, et al. (2016) Application of nanomedicine to the CNS diseases. Int Rev Neurobiol 130: 73-113.
  10. Zenaro E, Piacentino G, Constantin G (2016) The blood - brain barrier in Alzheimer's disease. Neurobiol Dis 107: 41-56.
  11. Qosa H, Mohamed A, Al Rihani SB, et al. (2016) High-throughput screening for identification of blood–brain barrier integrity enhancers: a drug repurposing opportunity to rectify vascular amyloid toxicity. J Alzheimers Dis 53: 1499-1516.
  12. Koizumi K, Wang G, Park L (2016) Endothelial dysfunction and amyloid-induced neurovascular alterations. Cell Mol Neurobiol 36: 155-165.
  13. Goldwaser EL, Acharya NK, Sarkar A, et al. (2016) Breakdown of the cerebrovasculature and blood-brain barrier: a mechanistic link between diabetes mellitus and Alzheimer's disease. J Alzheimers Dis 54: 445-456.
  14. Mahringer A, Reichel V, Ott M, et al. (2012) Overcoming the blood brain barrier: the challenge of brain drug targeting. J Nanoneurosci 2: 5-19.
  15. Fung KY, Wang C, Nyegaard S, et al. (2017) SR-BI mediated transcytosis of HDL in brain microvascular endothelial cells is independent of caveolin, clathrin, and PDZK1. Front Physiol 8: 841.
  16. Robert J, Button EB, Stukas S, et al. (2017) High-density lipoproteins suppress Aß-induced PBMC adhesion to human endothelial cells in bioengineered vessels and in monoculture. Mol Neurodegener 12: 60.
  17. Robert J, Stukas S, Button E, et al. (2016) Reconstituted high-density lipoproteins acutely reduce soluble brain Aß levels in symptomatic APP/PS1 mice. Biochim Biophys Acta 1862: 1027-1036.
  18. Hottman DA, Chernick D, Cheng S, et al. (2014) HDL and cognition in neurodegenerative disorders. Neurobiol Dis 72: 22-36.
  19. Weekman EM, Sudduth TL, Caverly CN, et al. (2016) Reduced efficacy of anti-A immunotherapy in a mouse model of amyloid deposition and vascular cognitive impairment comorbidity. J Neurosci 36: 9896-9907.
  20. Nelson AR, Sweeney MD, Sagare AP, et al. (2016) Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer's disease. Biochim Biophys Acta 1862: 887-900.
  21. Kapasi A, Schneider JA (2016) Vascular contributions to cognitive impairment, clinical Alzheimer's disease, and dementia in older persons. Biochim Biophys Acta 1862: 878-886.
  22. McAleese KL, Alafuzoff I, Charidimou A, et al. (2016) Post-mortem assessment in vascular dementia: Advances and aspirations. BMC Med 14: 129.
  23. Noh Y, Seo SW, Jeon S, et al. (2016) The role of cerebrovascular disease in amyloid deposition. J Alzheimers Dis 54: 1015-1026.
  24. Hishikawa N, Fukui Y, Sato K, et al. (2016) Cognitive and affective functions in Alzheimer's disease patients with metabolic syndrome. Eur J Neurol 23: 339-345.
  25. Gutierrez J, Honig L, Elkind MS, et al (2016) Brain arterial aging and its relationship to Alzheimer dementia. Neurology 86: 1507-1515.
  26. Nagata K, Yamazaki T, Takano D, et al. (2016) Cerebral circulation in aging. Ageing Res Rev 30: 49-60.
  27. Calabrese V, Giordano J, Signorile A, et al. (2016) Major pathogenic mechanisms in vascular dementia: roles of cellular stress response and hormesis in neuroprotection. J Neurosci Res 94: 1588-1603.
  28. Toth P, Tarantini S, Csiszar A, et al. (2017) 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 312: H1-H20.
  29. Devraj K, Poznanovic S, Spahn C, et al. (2016) 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 36: 1281-1294.
  30. Chao AC, Lee TC, Juo SH, et al. (2016) Hyperglycemia increases the production of amyloid-peptide leading to decreased endothelial tight junction. CNS Neurosci Ther 22: 291-297.
  31. Khalil RB, Khoury E, Koussa S (2016) Linking multiple pathogenic pathways in Alzheimer's disease. World J Psychiatry 6: 208-214.
  32. Festoff BW, Sajja RK, van Dreden P, et al. (2016) HGMB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer's disease. J Neuroinflamm 13: 194.
  33. Gangoda SV, Butlin M, Gupta V, et al. (2016) Pulsatile stretch alters expression and processing of amyloid precursor protein in human cerebral endothelial cells. J Hypertens 34: e24.
  34. Roberts AM, Jagadapillai R, Vaishnav RA, et al. (2016) Increased pulmonary arteriolar tone associated with lung oxidative stress and nitric oxide in a mouse model of Alzheimer's disease. Physiol Rep 4: e12953.
  35. Kyrtsos CR, Baras JS (2015) Modeling the role of the glymphatic pathway and cerebral blood vessel properties in Alzheimer's disease pathogenesis. PLoS One 10: e0139574.
  36. Kalaria RN, Akinyemi R, Ihara M (2016) Stroke injury, cognitive impairment and vascular dementia. Biochim Biophys Acta 1862: 915-925.
  37. Khan A, Kalaria RN, Corbett A, et al. (2016) Update on vascular dementia. J Geriatr Psychiatry Neurol 29: 281-301.
  38. Toda N, Okamura T (2016) Cigarette smoking impairs nitric oxide-mediated cerebral blood flow increase: implications for Alzheimer's disease. J Pharmacol Sci 131: 223-232.
  39. Uiterwijk R, Huijts M, Staals J, et al. (2016) Endothelial activation is associated with cognitive performance in patients with hypertension. Am J Hypertens 29: 464-469.
  40. Wang YJ (2014) Lessons from immunotherapy for Alzheimer's disease. Nat Rev Neurol 10: 188-189.
  41. Krstic D, Knuesel I (2013) Deciphering the mechanism underlying late-onset Alzheimer's disease. Nat Rev Neurol 9: 25-34.
  42. D'Arrigo JS (2011) Stable Nanoemulsions: Self-Assembly in Nature and Nanomedicine. Amsterdam: Elsevier, 415.
  43. Barbarese E, Ho SY, D'Arrigo JS, et al. (1995) Internalization of microbubbles by tumor cells in vivo and in vitro. J Neurooncol 26: 25-34.
  44. Garg G, Saraf SH, Saraf SW (2007) Cubosomes: an overview. Biol Pharm Bull 30: 350-353.
  45. (2007) Pouton CW Properties and uses of common formulation lipids, surfactants and cosurfactants. In: PP Constantinides, CJH Porter, Effective Utilization of Lipid-Based Systems to Enhance the Delivery of Poorly Soluble Drugs: Physicochemical, Biopharmaceutical and Product Development Considerations. Proceedings of the AAPSWorkshop, Arlington, VA, USA.
  46. Kaasgaard T, Drummond CJ (2006) Ordered 2-D and 3-D nano-structured amphiphile self-assembly materials stable in excess solvent. Phys Chem Chem Phys 8: 4957-4975.
  47. Small DM (1981) The behavior of biological lipids Pure Appl Chem 53: 2095-2103.
  48. Seddon JM, Robins J, Gulik-Krzywicki T, et al. (2000) Inverse micellar phases of phospholipids and glycolipids. Phys Chem Chem Phys 2: 4485-4493.
  49. Luzzati V, Vargas R, Mariani P, et al. (1993) Cubic phases of lipid-containing systems: elements of a theory and biological connotations. J Mol Biol 229: 540-551.
  50. Luzzati V, Vargas R, Gulik A, et al. (1992) Lipid polymorphism: a correction The structure of the cubic phase of extinction symbol Fd -consists of two types of disjointed reverse micelles embedded in a three-dimensional hydrocarbon matrix. Biochemistry 31: 279-285.
  51. D'Arrigo JS (1987) Surfactant mixtures, stable gas-in-liquid emulsions, and methods for the production of such emulsions from said mixtures.
  52. D'Arrigo JS (1993) Method for the production of medical-grade lipid-coated microbubbles, paramagnetic labeling of such microbubbles and therapeutic uses of microbubbles.
  53. Nimmrich V, Eckert A (2013) Calcium channel blockers and dementia. Brit J Pharmacol 169: 1203-1210.
  54. Shirwany NA, Payette D, Xie J, et al. (2007) The amyloid beta ion channel hypothesis of Alzheimer's disease. Neuropsychiatr Dis Treat 3: 597-612.
  55. Di Scala C, Yahi N, Boutemeur S, et al. (2016) Common molecular mechanism of amyloid pore formation by Alzheimer's ß-amyloid peptide and a-synuclein. Sci Rep 6: 28781.
  56. Demuro A, Smith M, Parker I (2011) Single-channel Ca2+ imaging implicates Aß1-42 amyloid pores in Alzheimer's disease pathology. J Cell Biol 195: 515-524.
  57. Serra-Batiste M, Ninot-Pedrosa M, Bayoumi M, et al. (2016) Aß42 assembles into specific ß-barrel pore-forming oligomers in membrane-mimicking environments. Proc Natl Acad Sci USA 113: 10866-10871.
  58. Bode DC, Baker MD, Viles JH (2017) Ion channel formation by amyloid-ß42 oligomers but not amyloid-ß40 in cellular membranes. J Biol Chem 292: 1404-1413.
  59. Wang GH, Jiang ZL, Li YC, et al. (2011) Free-radicle scavenger Edaravone treatment confers neuroprotection against traumatic brain injury in rats. J Neurotrauma 28: 2123-2134.
  60. Itoh T, Satou T, Nishida S, et al. (2010) Edaravone protects against apoptotic neuronal cell death and improves cerebral function after traumatic brain injury in rats. Neurochem Res 35: 348-355.
  61. Jiao SS, Yao XQ, Liu YH, et al. (2015) Edaravone alleviates Alzheimer's disease-type pathologies and cognitive deficits. Proc Natl Acad Sci USA 112: 5225-5230.
  62. Daulatzai MA (2016) Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer's disease. J Neurosci Res 95: 943-972.
  63. D'Arrigo JS (2020) Biomaterial to improve drug delivery in Alzheimer's disease: Linking major pathogenic pathways. OBM Geriatrics 4: 10.
  64. Gambini J (2020) Oxidative stress and inflammation: From mechanisms to therapeutic approaches Biomedicines.
  65. D'Arrigo JS (2020) Biomimetic nanocarrier targeting drug(s) to upstream-receptor mechanisms in dementia: Focusing on linking pathogenic cascades. Biomimetics 5: 11.
  66. Tangestani Fard M, Stough C (2019) A review and hypothesized model of the mechanisms that underpin the relationship between inflammation and cognition in the elderly. Front Aging Neurosci 11: 56.
  67. Khalil A, Berrougui H, Pawelec G, et al. (2012) Impairment of the ABCA1 and SR-BI-mediated cholesterol efflux pathways and HDL anti-inflammatory activity in Alzheimer's disease. Mech Ageing Dev 133: 20-29.
  68. Thanopoulou K, Fragkouli A, Stylianopoulou F, et al. (2010) Scavenger receptor class B type I (SR-BI) regulates perivascular macrophages and modifies amyloid pathology in an Alzheimer mouse model. Proc Natl Acad Sci 107: 20816-20821.
  69. Heringa SM, van den Ber E, Reijmer YD, et al. (2014) Markers of low-grade inflammation and endothelial dysfunction are related to reduced information processing speed and executive functioning in an older population -the Hoorn Study. Psychoneuroendocrinology 40: 108-118.
  70. D'Arrigo JS (2020) Nanotargeting dementia etiology: Aiming drug nanocarriers toward receptors for vascular endothelium, serum amyloid A, inflammasomes, and oxidative stress. Nano Prog 2: 25-30.
  71. Lenart N, Brough D, Denes A (2016) Inflammasomes link vascular disease with neuroinflammation and brain disorders. J Cereb Blood Flow Metab 36: 1668-1685.
  72. Sierksma A, Lu A, Mancuso R, et al. (2020) Novel Alzheimer risk genes determine the microglia response to amyloid-ß but not to TAU pathology. EMBO Mol Med 12: e10606.
  73. Lajoie JM, Shusta EV (2015) Targeting receptor-mediated transport for delivery of biologics across the blood-brain barrier .Annu Rev Pharmacol Toxicol 55: 613-631.
  74. Almer G, Mangge H, Zimmer A, et al. (2015) Lipoprotein-related and apolipoprotein-mediated delivery systems for drug targeting and imaging. Curr Med Chem 22: 3631-3651.
  75. Preston JE, Abbott J, Begley DJ (2014) Transcytosis of macromolecules at the blood-brain barrier. Adv Pharmacol 71: 147-163.
  76. Salmina AB, Inzhutova AI, Malinovskaya NA, et al. (2010) Endothelial dysfunction and repair in Alzheimer-type neurodegeneration: Neuronal and glial control. J Alzheimers Dis 22: 17-36.
  77. Tong XK, Hamel E (2015) Simvastatin restored vascular reactivity, endothelial function and reduced string vessel pathology in a mouse model of cerebrovascular disease. J Cereb Blood Flow Metab 35: 512-520.
  78. Koster KP, Thomas R, Morris AW, et al. (2016) Epidermal growth factor prevents oligomeric amyloid-induced angiogenesis deficits in vitro. J Cereb Blood Flow Metab 36: 1865-1871.
  79. Hostenbach S, D'haeseleer M, Kooijman R, et al. (2016) The pathophysiological role of astrocytic endothelin-1. Prog Neurobiol 144: 88-102.