Strategies for improving IPAD

R. Carare 2018, R.O.Carare@soton.ac.uk

Agents that improve contractility of vascular smooth muscle cells

Phosphodiesterase III is the major cAMP-hydrolyzing PDE uniquely expressed in vascular smooth muscle cells; PDE IIIA isoforms are also involved in cardiovascular function by regulating vascular smooth muscle growth and phenotypic changes. Cilostazol is a selective inhibitor of PDE III that increases cAMP in vascular cells and has multiple effects on the vasculature such as vasodilatation, anti-oxidation, anti-inflammation, regulation of smooth muscle cells, increase in cerebral haemodynamics and arterial elasticity with maintenance of microvascular integrity, as reviewed in (19). Cognition is significantly improved in experimental models and in humans receiving Cilostazol (20)(21)(22)(23). Administration of Cilostazol significantly improves IPAD and the brains of mice treated with Cilostazol show effects upon extracellular matrix, with upregulation of the anti-fibrillogenic glycoproteins (24)(25).

Using chaperones for efficient transport along the IPAD pathways

Clusterin (Apolipoprotein J) is a multifunctional protein that reduces the aggregation and toxicity of Aβ and appears to be beneficial in atherosclerosis (26)(27)We recently demonstrated that in APP/PS1 mouse models of Alzheimer’s disease, crossed with clusterin knockout mice, result in disappearance of Aβ plaques but an increase in severity of CAA. These findings suggest that clusterin is required for efficient chaperoning of solubilized proteins from plaques along IPAD (28). Administration of clusterin as a preventative therapy when the integrity and function of smooth muscle cells and basement membranes are not compromised may yield positive results for the prevention or delay in onset of symptoms of CAA and Alzheimer’s disease. Taxifolin is flavonoid that appears to maintain amyloid in its soluble forms more amenable for clearance (29) We are investigating whether Taxifolin facilitates IPAD.

Agents acting upon the innervation of smooth muscle cells

Experimental work is ongoing in this area. Results suggest that agents such as Prazosin, an alpha(1)-adrenoceptor antagonist, acting upon cholinergic or adrenergic innervation of cerebral arteries result in improvements of IPAD and in reduction of CAA in transgenic mouse models of Alzheimer’s disease (30).

 

references

1.
Kida S, Pantazis A, Weller R. CSF drains directly from the subarachnoid space into nasal lymphatics in the rat. Anatomy, histology and immunological significance. Neuropathol Appl Neurobiol. 1993;19(6):480-488. [PubMed]
2.
Bradbury M, Cserr H, Westrop R. Drainage of cerebral interstitial fluid into deep cervical lymph of the rabbit. Am J Physiol. 1981;240(4):F329-36. [PubMed]
3.
Szentistványi I, Patlak C, Ellis R, Cserr H. Drainage of interstitial fluid from different regions of rat brain. Am J Physiol. 1984;246(6 Pt 2):F835-44. [PubMed]
4.
Hawkes C, Härtig W, Kacza J, et al. Perivascular drainage of solutes is impaired in the ageing mouse brain and in the presence of cerebral amyloid angiopathy. Acta Neuropathol. 2011;121(4):431-443. [PubMed]
5.
Schley D, Carare-Nnadi R, Please CP, Perry VH, Weller RO. Mechanisms to explain the reverse perivascular transport of solutes out of the brain. J. 2006;238(4):962-974. doi: 10.1016/j.jtbi.2005.07.005
6.
Arbel-Ornath M, Hudry E, Eikermann-Haerter K, et al. Interstitial fluid drainage is impaired in ischemic stroke and Alzheimer’s disease mouse models. Acta Neuropathol. 2013;126(3):353-364. [PubMed]
7.
Diem A, MacGregor S, Gatherer M, Bressloff N, Carare R, Richardson G. Arterial Pulsations cannot Drive Intramural Periarterial Drainage: Significance for Aβ Drainage. Front Neurosci. 2017;11:475. [PMC]
8.
Weller R, Boche D, Nicoll J. Microvasculature changes and cerebral amyloid angiopathy in Alzheimer’s disease and their potential impact on therapy. Acta Neuropathol. 2009;118(1):87-102. [PubMed]
9.
Miners J, Van H, Chalmers K, Wilcock G, Love S, Kehoe P. Decreased expression and activity of neprilysin in Alzheimer disease are associated with cerebral amyloid angiopathy. J Neuropathol Exp Neurol. 2006;65(10):1012-1021. [PubMed]
10.
Zlokovic B. Clearing amyloid through the blood-brain barrier. J Neurochem. 2004;89(4):807-811. [PubMed]
11.
Iliff J, Wang M, Liao Y, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med. 2012;4(147):147ra111. [PubMed]
12.
Weller R, Djuanda E, Yow H, Carare R. Lymphatic drainage of the brain and the pathophysiology of neurological disease. Acta Neuropathol. 2009;117(1):1-14. [PubMed]
13.
Shibata M, Yamada S, Kumar S, et al. Clearance of Alzheimer’s amyloid-ss(1-40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest. 2000;106(12):1489-1499. [PubMed]
14.
Hawkes C, Jayakody N, Johnston D, Bechmann I, Carare R. Failure of perivascular drainage of β-amyloid in cerebral amyloid angiopathy. Brain Pathol. 2014;24(4):396-403. [PubMed]
15.
Hawkes C, Sullivan P, Hands S, Weller R, Nicoll J, Carare R. Disruption of Arterial Perivascular Drainage of Amyloid-β from the Brains of Mice Expressing the Human APOE ε4 Allele. PLoS One. 2012;7(7):e41636. [PMC]
16.
Tomic J, Pensalfini A, Head E, Glabe C. Soluble fibrillar oligomer levels are elevated in Alzheimer’s disease brain and correlate with cognitive dysfunction. Neurobiol Dis. 2009;35(3):352-358. [PubMed]
17.
Roher A, Kuo Y, Esh C, et al. Cortical and leptomeningeal cerebrovascular amyloid and white matter pathology in Alzheimer’s disease. Mol Med. 2003;9(3-4):112-122. [PubMed]
18.
Weller R, Hawkes C, Kalaria R, Werring D, Carare R. White matter changes in dementia: role of impaired drainage of interstitial fluid. Brain Pathol. 2015;25(1):63-78. [PubMed]
19.
Saito S, Ihara M. New therapeutic approaches for Alzheimer’s disease and cerebral amyloid angiopathy. Front Aging Neurosci. 2014 Oct 20;6:290. [PubMed]
20.
Saito S, Kojima S, Oishi N, Kakuta R, Maki T, Yasuno F, et al. A multicenter, randomized, placebo-controlled trial for cilostazol in patients with mild cognitive impairment: The COMCID study protocol. Alzheimers Dement (N Y). 2016 Oct 27;2(4):250–7. [PMC]
21.
Kitamura A, Manso Y, Duncombe J, Searcy J, Koudelka J, Binnie M, et al. Long-term cilostazol treatment reduces gliovascular damage and memory impairment in a mouse model of chronic cerebral hypoperfusion. Sci Rep. 2017 Jun 27;7(1):4299. [PubMed]
22.
Yanai S, Ito H, Endo S. Long-term cilostazol administration prevents age-related decline of hippocampus-dependent memory in mice. Neuropharmacology. 2018 Feb 1;129:57–68. [PubMed]
23.
Yanai S, Toyohara J, Ishiwata K, Ito H, Endo S. Long-term cilostazol administration ameliorates memory decline in senescence-accelerated mouse prone 8 (SAMP8) through a dual effect on cAMP and blood-brain barrier. Neuropharmacology. 2017 Apr 1;116:247–59. [PubMed]
24.
Maki T, Okamoto Y, Carare R, Hase Y, Hattori Y, Hawkes C, et al. Phosphodiesterase III inhibitor promotes drainage of cerebrovascular β-amyloid. Ann Clin Transl Neurol. 2014 Jul 8;1(8):519–33. [PMC]
25.
Manousopoulou A, Saito S, Yamamoto Y, Al-Daghri N, Ihara M, Carare R, et al. Hemisphere Asymmetry of Response to Pharmacologic Treatment in an Alzheimer’s Disease Mouse Model. J Alzheimers Dis. 2016 Mar 15;51(2):333–8. [PMC]
26.
Bielicki J, Zhang H, Cortez Y, Zheng Y, Narayanaswami V, Patel A, et al. A new HDL mimetic peptide that stimulates cellular cholesterol efflux with high efficiency greatly reduces atherosclerosis in mice. J Lipid Res. 2010 Jun 1;51(6):1496–503. [PubMed]
27.
Narayan P, Orte A, Clarke R, Bolognesi B, Hook S, Ganzinger K, et al. The extracellular chaperone clusterin sequesters oligomeric forms of the amyloid-β(1-40) peptide. Nat Struct Mol Biol. 2011 Dec 18;19(1):79–83. [PubMed]
28.
Wojtas AM, Kang SS, Olley BM, Gatherer M, Shinohara M, Lozano PA, et al. Loss of clusterin shifts amyloid deposition to the cerebrovasculature via disruption of perivascular drainage pathways. P [Internet]. 2017 Jul 12;114(33):E6962–71. Available from: http://dx.doi.org/10.1073/pnas.1701137114
29.
Saito S, Yamamoto Y, Maki T, Hattori Y, Ito H, Mizuno K, et al. Taxifolin inhibits amyloid-β oligomer formation and fully restores vascular integrity and memory in cerebral amyloid angiopathy. Acta Neuropathol Commun. 2017 Apr 4;5:26. [PMC]
30.
Katsouri L, Vizcaychipi M, McArthur S, Harrison I, Suárez-Calvet M, Lleo A, et al. Prazosin, an α(1)-adrenoceptor antagonist, prevents memory deterioration in the APP23 transgenic mouse model of Alzheimer’s disease. Neurobiol Aging. 2013 Apr 1;34(4):1105–15. [PubMed]