Interaction between AD and Cerebral Small Vessel Disease: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 1 by Si Eun Kim.

Alzheimer’s disease (AD) is characterized by the presence of β-amyloid (Aβ) and tau, and subcortical vascular cognitive impairment (SVCI) is characterized by cerebral small vessel disease (CSVD). They are the most common causes of cognitive impairment in the elderly population. Concurrent CSVD burden is more commonly observed in AD-type dementia than in other neurodegenerative diseases. Recent developments in Aβ and tau positron emission tomography (PET) have enabled the investigation of the relationship between AD biomarkers and CSVD in vivo.

  • subcortical vascular cognitive impairment
  • Alzheimer’s disease
  • ß-Amyloid
  • tau
  • cerebral small vessel disease
  • positron emission tomography

1. Introduction

Dementia is a progressive and deteriorative syndrome that affects memory and other cognitive domains, which interferes with a daily living [1]. Alzheimer’s disease (AD) and vascular dementia (VaD) are the two most common causes of dementia in the elderly [2]. AD is characterized by senile plaques formed by β-amyloid (Aβ) and neurofibrillary tangles (NFTs) formed by hyperphosphorylated tau. These changes, along with loss of neurons, contribute to the symptoms of dementia [3]. Based on these core AD pathological features, including Aβ [A], tau [T], and neurodegeneration [N] biomarkers, the National Institute on Aging—Alzheimer’s Association (NIA-AA) proposed the AT(N) classification system [4]. A and T biomarkers are specific for the Aβ plaques and tau NFTs that constitute the hallmark neuropathologic signs of AD, respectively, while biomarkers of (N) (such as atrophy on magnetic resonance imaging, MRI) are not disease specific [4,5][4][5]. Brain atrophy is indicative of the considerable loss of neurons and synapses in the cerebral cortex [6]. Although the assessment of atrophy lacks specificity to determine whether the cause is cell loss or synaptic loss, cortical thickness is widely used as a surrogate marker for neuronal loss [7,8][7][8]. The NIA-AA research framework defines AD biologically based on neuropathologic change or biomarkers and considers cognitive impairment a symptom or sign of the disease rather than the definition of the disease [4].
Further, Alzheimer’s disease is frequently associated with other aging-related disorders such as cerebrovascular disease, Lewy body disease, transactive response DNA-binding protein of 43 kDa (TDP-43) proteinopathies, and argyrophilic grain disease [6]. AD pathology mixed with vascular disease is more frequent in the elderly population (also known as mixed pathology in dementia or mixed dementia) [9]. Vascular diseases include arteriolosclerosis, cerebral amyloid angiopathy (CAA), atherosclerosis, macroinfarcts, and microinfarcts [10]. In fact, previous studies have shown that AD combined with vascular disease is the most prevalent mixed pathology [9,11,12][9][11][12].
Vascular dementia is caused by ischemic or hemorrhagic brain lesions that are characterized by numerous clinical syndromes [13]. The most common forms of VaD in the elderly are subcortical vascular dementia (SVaD), strategic infarct dementia, and multi-infarct dementia [14]. VaD is generally known to be the second most common cause of dementia in later life among Caucasian populations, although it may be the most common cause in East Asia [15,16,17][15][16][17]. SVaD, one of the main forms of VaD, is characterized by extensive cerebral small vessel disease (CSVD), including white matter hyperintensities (WMHs) and multiple lacunes [18]. Vascular risk factors, such as age, hypertension, and diabetes mellitus, contribute to the development of CSVD MRI markers. These markers gradually form deposits in subcortical regions over several decades, eventually resulting in SVaD [19]. Thus, SVaD shows a progression pattern similar to that of AD, which reveals an insidious onset and gradual progression; however, it is dissimilar to that of multi-infarct dementia (another major form of vascular dementia). From this perspective, there is a prodromal state of SVaD, referred to as subcortical vascular mild cognitive impairment (svMCI). Subcortical vascular cognitive impairment (SVCI), which incorporates SVaD and svMCI, refers to cognitive impairment caused by subcortical vascular lesions [20,21,22,23,24][20][21][22][23][24].

2. Frequency of AD Imaging Markers in SVCI

AD markers are more commonly observed in patients with SVCI than in cognitively unimpaired individuals. Specifically, in svMCI patients, the frequencies of Aβ positivity have been reported to be about 30% [25,29,49][25][26][27]. SVaD patients tend to display more frequent Aβ positivity than svMCI patients, ranging from 30% to 53% [25,50,51][25][28][29]. In terms of the tau marker, it has been shown that tau positivity is 70% (14/20) in Aβ (+) ADCI patients, 25.9% (7/27) in Aβ (+) SVCI patients, and 6.1% (2/33) in Aβ (−) SVCI patients. [52][30].

3. Correlation between AD and CSVD Imaging Markers

Molecular imaging studies have enabled uspeople to investigate the relationship between AD markers and CSVD MRI markers throughout the whole brain. There is increasing evidence from these studies showing that AD marker uptake is correlated with WMH volume, which is a characteristic MRI marker of CSVD. This has been observed prominently in the posterior regions of the brain. In ourthe previous study of 53 SVCI patients, a relationship between Aβ uptake and WMH volume was observed in APOE4 non-carriers [53][31]. WMH volume is correlated with Aβ uptake in the posterior cerebral regions. Another study using clustering analyses classified SVCI patients and AD patients into the Aβ occipital-predominant and Aβ occipital-sparing groups. The frequency of the occipital-predominant group has been shown to be higher in SVCI patients (62.2%) than in AD patients (37.8%) [33][32]. Furthermore, the Aβ spreading pattern in patients with SVCI is quite different from in patients with ADCI. Specifically, the Aβ spreading pattern of patients with SVCI demonstrates that Aβ accumulates in the occipital area before the temporal and frontal regions, whereas in patients with ADCI, the parietal and fronto-temporal regions precede the occipital region. [33,54,55,56,57][32][33][34][35][36]. The predominant Aβ deposition in the occipital region, mainly observed in patients with SVCI, may be related to the distribution pattern of CAA or ischemic vulnerability of the posterior circulation [53][31]. CAA is primarily found in the occipital region [53,58][31][37]. Moreover, ischemic injury and dysfunction of the endothelial layer may lead to disruption of the blood–brain barrier (BBB), which in turn leads to the deposition of Aβ. Since the vertebrobasilar system, which is responsible for the posterior circulation, may be vulnerable to ischemia, SVCI patients may show Aβ deposition primarily in the posterior region [53][31]. In terms of the relationship between CSVD and tau, previous studies have suggested that ischemia might increase tau burdens regardless of the amyloid pathway [63][38]. Animal studies have also shown an association between increased cerebrovascular pathology and tau formation [64][39]. In vivo imaging studies have shown that CSVD burden may be associated with higher tau accumulation in the inferior temporal regions regardless of Aβ positivity [65][40]. Furthermore, in terms of tau spreading order, patients with SVCI are quite different from patients with ADCI. Unlike in ADCI, tau accumulates earlier in the fusiform gyrus and inferior temporal gyrus than in the parahippocampal cortex in SVCI [65,66][40][41].

4. Potential Distinct Pathobiology of AD Markers in SVCI

Considering that SVCI and ADCI patients show different spreading patterns of AD imaging markers, there may be differences in the potential pathobiology of AD biomarkers between SVCI and ADCI patients. In patients with SVCI, vascular risk factors may lead to Aβ deposition. Several cohort studies have reported an association between vascular risk factors and Aβ deposition [67,68,69,70,71,72][42][43][44][45][46][47]. This Aβ deposition is increased by impaired Aβ clearance via a deficit in perivascular drainage of Aβ and breakdown of the BBB [73,74][48][49]. BBB breakdown causes faulty transport of Aβ through reduced levels of low-density lipoprotein receptor-related protein 1 (LRP1) and increased levels of receptor for advanced glycation end products (RAGE). These changes eventually lead to impaired clearance of toxic Aβ species [75,76][50][51]. Furthermore, Aβ accelerates the tau hyperphosphorylation by mediating the activation of protein kinases, including cyclin-dependent kinase 5 (CDK-5) and glycogen synthase kinase 3β (GSK-3β) [77,78][52][53]. In addition, Aβ induces the activation of caspase-3 and calpain-1 and the cleavage of tau, generating neurotoxic tau fragments [79,80][54][55]. The link between Aβ and tau aggregation may involve microglial activation [81][56]. Soluble Aβ oligomers are known to activate microglial cells [82][57]. Mouse studies on transgenic AD have revealed that the microglial activation precedes tau aggregation [83][58] and facilitates tau hyperphosphorylation through cytokine release with subsequent NFT formation [84][59]. There are two potential mechanisms that may explain how vascular risk factors induce tau accumulation. One hypothesis is that ischemia may activate CDK-5 and GSK-3β, resulting in tau phosphorylation [85][60]. Activation of CDK-5 occurs when ischemia inhibits the pumping of calcium ions out of cells and raises intracellular calcium levels [86,87][61][62]. GSK-3β is activated by ischemia through decreased activity of the phosphatidylinositol 3-kinase/Akt pathway [88,89][63][64]. Moreover, vascular risk factors and the accumulation of Aβ plaques lead to oxidative stress [90,91,92][65][66][67]. Oxidative stress may also be caused by several mechanisms, such as mitochondrial dysfunction or inflammatory responses [92][67]. It may manifest as damage to synapses and changes in Ca2+ homeostasis, resulting in an apoptotic cascade and neurotoxicity [92][67]. Notably, there are distinct effects of APOE genotyping on Aβ deposition between patients with SVCI and ADCI. Specifically, apolipoprotein E4 (APOE4) is a risk factor for Aβ positivity in patients with ADCI and SVCI. Apolipoprotein E2 (APOE2) is a protective factor in ADCI (OR = 0.43); however, it is a risk factor in SVCI (OR = 2.26) [93][68]. Thus, APOE2 might accelerate apolipoprotein E leakage in the vessel walls of patients with SVCI, which in turn leads to impaired vascular drainage of Aβ. This impaired drainage eventually results in increased Aβ burdens in the brain parenchyma [93][68]. Alternatively, APOE2 may contribute to the development of CAA, which in turn leads to increased CSVD [93][68].

5. Clinical Effects of AD and CSVD Markers in SVCI Patients

There has been some debate related to the clinical effects of Aβ and CSVD imaging markers. In fact, among patients with extensive WMHs, some tend to show severe dementia symptoms, while others have no symptoms. In this regard, ourthe previous studies investigated which imaging markers might affect the clinical features of SVCI and found that AD biomarkers and CSVD independently affect cognition, abnormal behavior, and gait disturbances [29,32,51,65,94,95,96][26][29][40][69][70][71][72]. A cross-sectional study has reported that Aβ uptake is only associated with memory dysfunction, whereas CSVD burden is associated with memory, visuospatial, and frontal executive functions [94][70]. Longitudinal cohort studies have also shown that Aβ positivity is associated with faster cognitive decline in patients with SVaD [51][29] and higher conversion to dementia in patients with svMCI [32][69]. In terms of abnormal behavior, Aβ predicts the signs of delusions and irritability, while CSVD burdens are associated with other behavioral symptoms, such as apathy and depression [97][73]. In addition, periventricular WMHs are the most important predictor of gait disturbances [98][74]. SVCI patients show distinct brain structural and cognitive trajectories based on AT (Aβ/tau) biomarker profiles [52][30]. A previous study showed that patients in the A+T+ group predicted a more rapid decline in structural and cognitive trajectories than those in the A+T− group, followed by those in the A−T− group [52][30]. Moreover, AD markers and CSVD burden have a synergistic effect on cognitive decline. In a cross-sectional study, significant interactions between WMHs and Aβ uptake were apparent in visuospatial function, suggesting that CSVD and Aβ synergistically affect cognitive impairment [29][26]. A longitudinal study comparing patients with SVCI and ADCI who had similar tau levels has shown that as Aβ turns positive, SVCI shows a steeper cognitive decline compared to the ADCI group [99][75]. In addition, as tau levels increase, the SVCI group shows a steeper cognitive decline than the ADCI group [99][75]. These findings indicate that there are interactive effects between AD markers and CSVD on cognitive decline.
Furthermore, Aβ and CSVD affect specific downstream imaging markers, such as network changes and brain atrophy in specific regions, which in turn lead to the development of these corresponding clinical outcomes [32,95,100][69][71][76]. Specifically, Aβ uptake is associated with cortical thinning in the medial temporal regions including hippocampal changes, which in turn leads to memory dysfunction. In contrast, CSVD burdens are primarily associated with frontal thinning [101][77] and white matter network disruption [95][71], which in turn leads to frontal dysfunction. In addition, a three-year longitudinal study has shown that time-varying Aβ and CSVD affects the temporoparietal and frontal thinning, respectively, which in turn contributes to the corresponding cognitive decline [32][69]. Another cross-sectional study has demonstrated that Aβ positivity and CSVD severity are independently associated with higher tau uptake in the medial and inferior temporal regions, respectively [65][40]. Moreover, increased tau uptake can mediate the relationship between Aβ and CSVD uptake and cognitive impairment, indicating that tau is another important common downstream marker of Aβ and CSVD burdens.

6. Hemorrhagic Markers in Cerebral Amyloid Angiopathy (CAA) and the Clinical Effects

CAA is characterized by Aβ deposition in the small arteries of the meninges and cortex, which causes vascular dysfunction and brain injury [102][78]. CAA is clinically and radiologically characterized by lobar intracerebral hemorrhage (ICH), strictly lobar cerebral microbleeds (CMBs) and cortical superficial siderosis (CSS) [103][79]. CAA is generally related to Aβ parenchymal aggregates, such as neuritic and diffuse plaques, although it can also occur pathologically without evident AD neuropathological changes [104][80]. Generally, the anatomical location of CMBs reflects their underlying etiology. Specifically, deep CMBs are presumed to be due to hypertensive CSVD, whereas lobar CMBs may reflect CAA [105][81]. In a cross-sectional study, consistent with previous studies, Aβ uptake has been shown to be associated with lobar CMBs [23]. CSVD is also associated with lobar CMBs as well as deep CMBs [23]. Aβ uptake and CSVD synergistically affect the development of lobar CMB [23]. Furthermore, a longitudinal study demonstrated that longitudinal measures of Aβ uptake and lacunes synergistically affect the development of lobar CMBs [69][44]. According to Thal’s CAA pathologic stage, CAA pathology extends sequentially from leptomeningeal and cortical vessels to cerebellar vessels and eventually to the striatum and brainstem vessels [106][82]. Patients with both cerebellar and lobar CMBs are more likely to present with CAA features, whereas deep CMBs, regardless of the presence of both lobar and cerebellar CMBs, are more likely to represent underlying hypertensive angiopathy than CAA features [107][83]. Interestingly, restricted superficial cerebellar CMBs refer to CAA imaging markers, whereas the involvement of the cerebellar dentate nucleus might be equivalent to deep CMB [107][83]. OurThe previous study showed that the frequency of APOE4 was higher in Aβ (+) CAA than in Aβ (−) CAA, whereas APOE2 was associated with overt hemorrhagic markers of CAA, such as lobar ICH and CSS [108][84]. These findings are consistent with other studies showing that APOE4 is related to the deposition of Aβ burdens [109[85][86],110], and APOE 2 is related to the breakdown of blood vessel walls [111][87]. In addition, the number of lobar CMB and the presence of CSS can predict Aβ (+), whereas ischemic CSVD markers can predict Aβ (−) [108][84].

References

  1. Scott, K.R.; Barrett, A.M. Dementia syndromes: Evaluation and treatment. Expert Rev. Neurother. 2007, 7, 407–422.
  2. Roman, G.C. Vascular dementia: Distinguishing characteristics, treatment, and prevention. J. Am. Geriatr. Soc. 2003, 51 (Suppl. S5), S296–S304.
  3. Wenk, G.L. Neuropathologic changes in Alzheimer’s disease. J. Clin. Psychiatry 2003, 64 (Suppl. 9), 7–10.
  4. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2018, 14, 535–562.
  5. Jack, C.R., Jr.; Wiste, H.J.; Therneau, T.M.; Weigand, S.D.; Knopman, D.S.; Mielke, M.M.; Lowe, V.J.; Vemuri, P.; Machulda, M.M.; Schwarz, C.G.; et al. Associations of Amyloid, Tau, and Neurodegeneration Biomarker Profiles With Rates of Memory Decline Among Individuals Without Dementia. Jama 2019, 321, 2316–2325.
  6. DeTure, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32.
  7. Lerch, J.P.; Pruessner, J.C.; Zijdenbos, A.; Hampel, H.; Teipel, S.J.; Evans, A.C. Focal decline of cortical thickness in Alzheimer’s disease identified by computational neuroanatomy. Cereb. Cortex 2005, 15, 995–1001.
  8. Márquez, F.; Yassa, M.A. Neuroimaging Biomarkers for Alzheimer’s Disease. Mol. Neurodegener. 2019, 14, 21.
  9. Schneider, J.A.; Arvanitakis, Z.; Bang, W.; Bennett, D.A. Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology 2007, 69, 2197–2204.
  10. Agrawal, S.; Schneider, J.A. Vascular pathology and pathogenesis of cognitive impairment and dementia in older adults. Cereb. Circ.-Cogn. Behav. 2022, 3, 100148.
  11. Attems, J.; Jellinger, K.A. The overlap between vascular disease and Alzheimer’s disease--lessons from pathology. BMC Med. 2014, 12, 206.
  12. Boyle, P.A.; Yu, L.; Nag, S.; Leurgans, S.; Wilson, R.S.; Bennett, D.A.; Schneider, J.A. Cerebral amyloid angiopathy and cognitive outcomes in community-based older persons. Neurology 2015, 85, 1930–1936.
  13. Gorelick, P.B.; Scuteri, A.; Black, S.E.; Decarli, C.; Greenberg, S.M.; Iadecola, C.; Launer, L.J.; Laurent, S.; Lopez, O.L.; Nyenhuis, D.; et al. Vascular contributions to cognitive impairment and dementia: A statement for healthcare professionals from the american heart association/american stroke association. Stroke 2011, 42, 2672–2713.
  14. Thal, D.R.; Grinberg, L.T.; Attems, J. Vascular dementia: Different forms of vessel disorders contribute to the development of dementia in the elderly brain. Exp. Gerontol. 2012, 47, 816–824.
  15. Jorm, A.F.; Jolley, D. The incidence of dementia: A meta-analysis. Neurology 1998, 51, 728–733.
  16. Lobo, A.; Launer, L.J.; Fratiglioni, L.; Andersen, K.; Di Carlo, A.; Breteler, M.M.; Copeland, J.R.; Dartigues, J.F.; Jagger, C.; Martinez-Lage, J.; et al. Prevalence of dementia and major subtypes in Europe: A collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group. Neurology 2000, 54 (Suppl. 5), S4–S9.
  17. Iadecola, C.; Duering, M.; Hachinski, V.; Joutel, A.; Pendlebury, S.T.; Schneider, J.A.; Dichgans, M. Vascular Cognitive Impairment and Dementia: JACC Scientific Expert Panel. J. Am. Coll. Cardiol. 2019, 73, 3326–3344.
  18. O’Brien, J.T.; Erkinjuntti, T.; Reisberg, B.; Roman, G.; Sawada, T.; Pantoni, L.; Bowler, J.V.; Ballard, C.; DeCarli, C.; Gorelick, P.B.; et al. Vascular cognitive impairment. Lancet Neurol 2003, 2, 89–98.
  19. Roman, G.C.; Erkinjuntti, T.; Wallin, A.; Pantoni, L.; Chui, H.C. Subcortical ischaemic vascular dementia. Lancet Neurol. 2002, 1, 426–436.
  20. Seo, S.W.; Cho, S.S.; Park, A.; Chin, J.; Na, D.L. Subcortical vascular versus amnestic mild cognitive impairment: Comparison of cerebral glucose metabolism. J. Neuroimaging 2009, 19, 213–219.
  21. Seo, S.W.; Ahn, J.; Yoon, U.; Im, K.; Lee, J.M.; Tae Kim, S.; Ahn, H.J.; Chin, J.; Jeong, Y.; Na, D.L. Cortical thinning in vascular mild cognitive impairment and vascular dementia of subcortical type. J. Neuroimaging 2010, 20, 37–45.
  22. Seo, S.W.; Im, K.; Lee, J.M.; Kim, Y.H.; Kim, S.T.; Kim, S.Y.; Yang, D.W.; Kim, S.I.; Cho, Y.S.; Na, D.L. Cortical thickness in single- versus multiple-domain amnestic mild cognitive impairment. Neuroimage 2007, 36, 289–297.
  23. Park, J.H.; Seo, S.W.; Kim, C.; Kim, G.H.; Noh, H.J.; Kim, S.T.; Kwak, K.C.; Yoon, U.; Lee, J.M.; Lee, J.W.; et al. Pathogenesis of cerebral microbleeds: In vivo imaging of amyloid and subcortical ischemic small vessel disease in 226 individuals with cognitive impairment. Ann. Neurol. 2013, 73, 584–593.
  24. Dubois, M.F.; Hebert, R. The incidence of vascular dementia in Canada: A comparison with Europe and East Asia. Neuroepidemiology 2001, 20, 179–187.
  25. Kang, S.H.; Kim, M.E.; Jang, H.; Kwon, H.; Lee, H.; Kim, H.J.; Seo, S.W.; Na, D.L. Amyloid Positivity in the Alzheimer/Subcortical-Vascular Spectrum. Neurology 2021, 96, e2201–e2211.
  26. Lee, M.J.; Seo, S.W.; Na, D.L.; Kim, C.; Park, J.H.; Kim, G.H.; Kim, C.H.; Noh, Y.; Cho, H.; Kim, H.J.; et al. Synergistic effects of ischemia and beta-amyloid burden on cognitive decline in patients with subcortical vascular mild cognitive impairment. JAMA Psychiatry 2014, 71, 412–422.
  27. Chong, J.S.X.; Jang, H.; Kim, H.J.; Ng, K.K.; Na, D.L.; Lee, J.H.; Seo, S.W.; Zhou, J. Amyloid and cerebrovascular burden divergently influence brain functional network changes over time. Neurology 2019, 93, e1514–e1525.
  28. Lee, J.H.; Kim, S.H.; Kim, G.H.; Seo, S.W.; Park, H.K.; Oh, S.J.; Kim, J.S.; Cheong, H.K.; Na, D.L. Identification of pure subcortical vascular dementia using 11C-Pittsburgh compound B. Neurology 2011, 77, 18–25.
  29. Ye, B.S.; Seo, S.W.; Kim, J.H.; Kim, G.H.; Cho, H.; Noh, Y.; Kim, H.J.; Yoon, C.W.; Woo, S.Y.; Kim, S.H.; et al. Effects of amyloid and vascular markers on cognitive decline in subcortical vascular dementia. Neurology 2015, 85, 1687–1693.
  30. Jang, H.; Kim, H.J.; Park, S.; Park, Y.H.; Choe, Y.; Cho, H.; Lyoo, C.H.; Yoon, U.; Lee, J.S.; Kim, Y.; et al. Application of an amyloid and tau classification system in subcortical vascular cognitive impairment patients. Eur. J. Nucl. Med. Mol. Imaging 2020, 47, 292–303.
  31. Noh, Y.; Seo, S.W.; Jeon, S.; Lee, J.M.; Kim, J.H.; Kim, G.H.; Cho, H.; Yoon, C.W.; Kim, H.J.; Ye, B.S.; et al. White matter hyperintensities are associated with amyloid burden in APOE4 non-carriers. J. Alzheimers Dis. 2014, 40, 877–886.
  32. Jang, H.; Park, J.Y.; Jang, Y.K.; Kim, H.J.; Lee, J.S.; Na, D.L.; Noh, Y.; Lockhart, S.N.; Seong, J.K.; Seo, S.W. Distinct amyloid distribution patterns in amyloid positive subcortical vascular cognitive impairment. Sci. Rep. 2018, 8, 16178.
  33. Lehmann, M.; Ghosh, P.M.; Madison, C.; Laforce, R., Jr.; Corbetta-Rastelli, C.; Weiner, M.W.; Greicius, M.D.; Seeley, W.W.; Gorno-Tempini, M.L.; Rosen, H.J.; et al. Diverging patterns of amyloid deposition and hypometabolism in clinical variants of probable Alzheimer’s disease. Brain A J. Neurol. 2013, 136 Pt 3, 844–858.
  34. Jang, H.; Seo, S.W. Comparison of Amyloid and Tau Patterns and Neuropsychological Findings in Cerebral Amyloid Angiopathy and Hypertensive Small Vessel Disease. In Proceedings of the Conference of Korean Dementia Association, Seoul, Korea, 4 November 2017; Korea Dementia Association: Seoul, Korea, 2017.
  35. Grothe, M.J.; Barthel, H.; Sepulcre, J.; Dyrba, M.; Sabri, O.; Teipel, S.J. In vivo staging of regional amyloid deposition. Neurology 2017, 89, 2031–2038.
  36. van der Kant, R.; Goldstein, L.S.B.; Ossenkoppele, R. Amyloid-β-independent regulators of tau pathology in Alzheimer disease. Nat. Rev. Neurosci. 2020, 21, 21–35.
  37. Viswanathan, A.; Greenberg, S.M. Cerebral amyloid angiopathy in the elderly. Ann. Neurol. 2011, 70, 871–880.
  38. Song, B.; Ao, Q.; Wang, Z.; Liu, W.; Niu, Y.; Shen, Q.; Zuo, H.; Zhang, X.; Gong, Y. Phosphorylation of tau protein over time in rats subjected to transient brain ischemia. Neural. Regen. Res. 2013, 8, 3173–3182.
  39. Bennett, R.E.; Robbins, A.B.; Hu, M.; Cao, X.; Betensky, R.A.; Clark, T.; Das, S.; Hyman, B.T. Tau induces blood vessel abnormalities and angiogenesis-related gene expression in P301L transgenic mice and human Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2018, 115, E1289–E1298.
  40. Kim, H.J.; Park, S.; Cho, H.; Jang, Y.K.; San Lee, J.; Jang, H.; Kim, Y.; Kim, K.W.; Ryu, Y.H.; Choi, J.Y.; et al. Assessment of Extent and Role of Tau in Subcortical Vascular Cognitive Impairment Using 18F-AV1451 Positron Emission Tomography Imaging. JAMA Neurol. 2018, 75, 999–1007.
  41. Cho, H.; Choi, J.Y.; Hwang, M.S.; Kim, Y.J.; Lee, H.M.; Lee, H.S.; Lee, J.H.; Ryu, Y.H.; Lee, M.S.; Lyoo, C.H. In vivo cortical spreading pattern of tau and amyloid in the Alzheimer disease spectrum. Ann. Neurol. 2016, 80, 247–258.
  42. Gottesman, R.F.; Schneider, A.L.; Zhou, Y.; Coresh, J.; Green, E.; Gupta, N.; Knopman, D.S.; Mintz, A.; Rahmim, A.; Sharrett, A.R.; et al. Association Between Midlife Vascular Risk Factors and Estimated Brain Amyloid Deposition. JAMA 2017, 317, 1443–1450.
  43. Hughes, T.M.; Wagenknecht, L.E.; Craft, S.; Mintz, A.; Heiss, G.; Palta, P.; Wong, D.; Zhou, Y.; Knopman, D.; Mosley, T.H.; et al. Arterial stiffness and dementia pathology: Atherosclerosis Risk in Communities (ARIC)-PET Study. Neurology 2018, 90, e1248–e1256.
  44. Rabin, J.S.; Schultz, A.P.; Hedden, T.; Viswanathan, A.; Marshall, G.A.; Kilpatrick, E.; Klein, H.; Buckley, R.F.; Yang, H.S.; Properzi, M.; et al. Interactive Associations of Vascular Risk and beta-Amyloid Burden with Cognitive Decline in Clinically Normal Elderly Individuals: Findings From the Harvard Aging Brain Study. JAMA Neurol. 2018, 75, 1124–1131.
  45. Arfanakis, K.; Evia, A.M.; Leurgans, S.E.; Cardoso, L.F.C.; Kulkarni, A.; Alqam, N.; Lopes, L.F.; Vieira, D.; Bennett, D.A.; Schneider, J.A. Neuropathologic Correlates of White Matter Hyperintensities in a Community-Based Cohort of Older Adults. J. Alzheimers Dis. 2020, 73, 333–345.
  46. Köbe, T.; Gonneaud, J.; Pichet Binette, A.; Meyer, P.F.; McSweeney, M.; Rosa-Neto, P.; Breitner, J.C.S.; Poirier, J.; Villeneuve, S. Association of Vascular Risk Factors with β-Amyloid Peptide and Tau Burdens in Cognitively Unimpaired Individuals and Its Interaction with Vascular Medication Use. JAMA Netw. Open. 2020, 3, e1920780.
  47. Lockhart, S.N.; Schaich, C.L.; Craft, S.; Sachs, B.C.; Rapp, S.R.; Jung, Y.; Whitlow, C.T.; Solingapuram Sai, K.K.; Cleveland, M.; Williams, B.J.; et al. Associations among vascular risk factors, neuroimaging biomarkers, and cognition: Preliminary analyses from the Multi-Ethnic Study of Atherosclerosis (MESA). Alzheimers Dement. 2022, 18, 551–560.
  48. Charidimou, A.; Boulouis, G.; Gurol, M.E.; Ayata, C.; Bacskai, B.J.; Frosch, M.P.; Viswanathan, A.; Greenberg, S.M. Emerging concepts in sporadic cerebral amyloid angiopathy. Brain A J. Neurol. 2017, 140, 1829–1850.
  49. Grimmer, T.; Faust, M.; Auer, F.; Alexopoulos, P.; Forstl, H.; Henriksen, G.; Perneczky, R.; Sorg, C.; Yousefi, B.H.; Drzezga, A.; et al. White matter hyperintensities predict amyloid increase in Alzheimer’s disease. Neurobiol. Aging 2012, 33, 2766–2773.
  50. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150.
  51. Montagne, A.; Zhao, Z.; Zlokovic, B.V. Alzheimer’s disease: A matter of blood-brain barrier dysfunction? J. Exp. Med. 2017, 214, 3151–3169.
  52. Terwel, D.; Muyllaert, D.; Dewachter, I.; Borghgraef, P.; Croes, S.; Devijver, H.; Van Leuven, F. Amyloid activates GSK-3beta to aggravate neuronal tauopathy in bigenic mice. Am. J. Pathol. 2008, 172, 786–798.
  53. Hernandez, P.; Lee, G.; Sjoberg, M.; Maccioni, R.B. Tau phosphorylation by cdk5 and Fyn in response to amyloid peptide Abeta (25-35): Involvement of lipid rafts. J. Alzheimers Dis. 2009, 16, 149–156.
  54. Gamblin, T.C.; Chen, F.; Zambrano, A.; Abraha, A.; Lagalwar, S.; Guillozet, A.L.; Lu, M.; Fu, Y.; Garcia-Sierra, F.; LaPointe, N.; et al. Caspase cleavage of tau: Linking amyloid and neurofibrillary tangles in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2003, 100, 10032–10037.
  55. Park, S.-Y.; Ferreira, A. The Generation of a 17 kDa Neurotoxic Fragment: An Alternative Mechanism by which Tau Mediates β-Amyloid-Induced Neurodegeneration. J. Neurosci. 2005, 25, 5365–5375.
  56. Ismail, R.; Parbo, P.; Madsen, L.S.; Hansen, A.K.; Hansen, K.V.; Schaldemose, J.L.; Kjeldsen, P.L.; Stokholm, M.G.; Gottrup, H.; Eskildsen, S.F.; et al. The relationships between neuroinflammation, beta-amyloid and tau deposition in Alzheimer’s disease: A longitudinal PET study. J. Neuroinflamm. 2020, 17, 151.
  57. Heneka, M.T.; Carson, M.J.; Khoury, J.E.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405.
  58. Yoshiyama, Y.; Higuchi, M.; Zhang, B.; Huang, S.M.; Iwata, N.; Saido, T.C.; Maeda, J.; Suhara, T.; Trojanowski, J.Q.; Lee, V.M. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 2007, 53, 337–351.
  59. Maphis, N.; Xu, G.; Kokiko-Cochran, O.N.; Jiang, S.; Cardona, A.; Ransohoff, R.M.; Lamb, B.T.; Bhaskar, K. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain A J. Neurol. 2015, 138 Pt 6, 1738–1755.
  60. Korte, N.; Nortley, R.; Attwell, D. Cerebral blood flow decrease as an early pathological mechanism in Alzheimer’s disease. Acta Neuropathol. 2020, 140, 793–810.
  61. Liu, S.L.; Wang, C.; Jiang, T.; Tan, L.; Xing, A.; Yu, J.T. The Role of Cdk5 in Alzheimer’s Disease. Mol. Neurobiol. 2016, 53, 4328–4342.
  62. Toro-Fernández, L.F.; Zuluaga-Monares, J.C.; Saldarriaga-Cartagena, A.M.; Cardona-Gómez, G.P.; Posada-Duque, R. Targeting CDK5 in Astrocytes Promotes Calcium Homeostasis Under Excitotoxic Conditions. Front. Cell. Neurosci. 2021, 15, 643717.
  63. Mottet, D.; Dumont, V.; Deccache, Y.; Demazy, C.; Ninane, N.; Raes, M.; Michiels, C. Regulation of hypoxia-inducible factor-1alpha protein level during hypoxic conditions by the phosphatidylinositol 3-kinase/Akt/glycogen synthase kinase 3beta pathway in HepG2 cells. J. Biol. Chem. 2003, 278, 31277–31285.
  64. Curtis, D.; Bandyopadhyay, S. Mini-review: Role of the PI3K/Akt pathway and tyrosine phosphatases in Alzheimer’s disease susceptibility. Ann. Hum. Genet. 2021, 85, 1–6.
  65. Grochowski, C.; Litak, J.; Kamieniak, P.; Maciejewski, R. Oxidative stress in cerebral small vessel disease. Role of reactive species. Free Radic. Res. 2018, 52, 1–13.
  66. Hort, J.; Vališ, M.; Kuča, K.; Angelucci, F. Vascular Cognitive Impairment: Information from Animal Models on the Pathogenic Mechanisms of Cognitive Deficits. Int. J. Mol. Sci. 2019, 20, 2405.
  67. Yuste, J.E.; Tarragon, E.; Campuzano, C.M.; Ros-Bernal, F. Implications of glial nitric oxide in neurodegenerative diseases. Front. Cell. Neurosci. 2015, 9, 322.
  68. Lee, J.S.; Lee, H.; Park, S.; Choe, Y.; Park, Y.H.; Cheon, B.K.; Hahn, A.; Ossenkoppele, R.; Kim, H.J.; Kim, S.; et al. Association between APOE epsilon2 and Abeta burden in patients with Alzheimer- and vascular-type cognitive impairment. Neurology 2020, 95, e2354–e2365.
  69. Kim, H.J.; Yang, J.J.; Kwon, H.; Kim, C.; Lee, J.M.; Chun, P.; Kim, Y.J.; Jung, N.Y.; Chin, J.; Kim, S.; et al. Relative impact of amyloid-beta, lacunes, and downstream imaging markers on cognitive trajectories. Brain 2016, 139 Pt 9, 2516–2527.
  70. Park, J.H.; Seo, S.W.; Kim, C.; Kim, S.H.; Kim, G.H.; Kim, S.T.; Jeon, S.; Lee, J.M.; Oh, S.J.; Kim, J.S.; et al. Effects of cerebrovascular disease and amyloid beta burden on cognition in subjects with subcortical vascular cognitive impairment. Neurobiol. Aging 2014, 35, 254–260.
  71. Kim, H.J.; Im, K.; Kwon, H.; Lee, J.M.; Kim, C.; Kim, Y.J.; Jung, N.Y.; Cho, H.; Ye, B.S.; Noh, Y.; et al. Clinical effect of white matter network disruption related to amyloid and small vessel disease. Neurology 2015, 85, 63–70.
  72. Jang, Y.K.; Kwon, H.; Kim, Y.J.; Jung, N.Y.; Lee, J.S.; Lee, J.; Chin, J.; Im, K.; Jeon, S.; Lee, J.M.; et al. Early- vs late-onset subcortical vascular cognitive impairment. Neurology 2016, 86, 527–534.
  73. Kim, H.J.; Ye, B.S.; Yoon, C.W.; Cho, H.; Noh, Y.; Kim, G.H.; Choi, Y.S.; Kim, J.H.; Jeon, S.; Lee, J.M.; et al. Effects of APOE epsilon4 on brain amyloid, lacunar infarcts, and white matter lesions: A study among patients with subcortical vascular cognitive impairment. Neurobiol. Aging 2013, 34, 2482–2487.
  74. Kim, Y.J.; Kwon, H.K.; Lee, J.M.; Cho, H.; Kim, H.J.; Park, H.K.; Jung, N.Y.; San Lee, J.; Lee, J.; Jang, Y.K.; et al. Gray and white matter changes linking cerebral small vessel disease to gait disturbances. Neurology 2016, 86, 1199–1207.
  75. Jang, H.; Kim, H.J.; Choe, Y.S.; Kim, S.J.; Park, S.; Kim, Y.; Kim, K.W.; Lyoo, C.H.; Cho, H.; Ryu, Y.H.; et al. The Impact of Amyloid-beta or Tau on Cognitive Change in the Presence of Severe Cerebrovascular Disease. J. Alzheimers Dis. 2020, 78, 573–585.
  76. Byeon, H. A Prediction Model for Mild Cognitive Impairment Using Random Forests. Int. J. Adv. Comput. Sci. Appl. (IJACSA) 2015, 6, 1–5.
  77. Ye, B.S.; Seo, S.W.; Kim, G.H.; Noh, Y.; Cho, H.; Yoon, C.W.; Kim, H.J.; Chin, J.; Jeon, S.; Lee, J.M.; et al. Amyloid burden, cerebrovascular disease, brain atrophy, and cognition in cognitively impaired patients. Alzheimers Dement. 2015, 11, 494–503.e3.
  78. Weller, R.O.; Massey, A.; Newman, T.A.; Hutchings, M.; Kuo, Y.M.; Roher, A.E. Cerebral amyloid angiopathy: Amyloid beta accumulates in putative interstitial fluid drainage pathways in Alzheimer’s disease. Am. J. Pathol. 1998, 153, 725–733.
  79. Yamada, M. Cerebral amyloid angiopathy: Emerging concepts. J. Stroke 2015, 17, 17–30.
  80. Greenberg, S.M.; Bacskai, B.J.; Hernandez-Guillamon, M.; Pruzin, J.; Sperling, R.; van Veluw, S.J. Cerebral amyloid angiopathy and Alzheimer disease—one peptide, two pathways. Nat. Rev. Neurol. 2020, 16, 30–42.
  81. Greenberg, S.M.; Vernooij, M.W.; Cordonnier, C.; Viswanathan, A.; Al-Shahi Salman, R.; Warach, S.; Launer, L.J.; Van Buchem, M.A.; Breteler, M.M.; Microbleed Study, G. Cerebral microbleeds: A guide to detection and interpretation. Lancet Neurol. 2009, 8, 165–174.
  82. Thal, D.R.; Ghebremedhin, E.; Orantes, M.; Wiestler, O.D. Vascular pathology in Alzheimer disease: Correlation of cerebral amyloid angiopathy and arteriosclerosis/lipohyalinosis with cognitive decline. J. Neuropathol. Exp. Neurol. 2003, 62, 1287–1301.
  83. Jung, Y.H.; Jang, H.; Park, S.B.; Choe, Y.S.; Park, Y.; Kang, S.H.; Lee, J.M.; Kim, J.S.; Kim, J.; Kim, J.P.; et al. Strictly Lobar Microbleeds Reflect Amyloid Angiopathy Regardless of Cerebral and Cerebellar Compartments. Stroke 2020, 51, 3600–3607.
  84. Jang, H.; Jang, Y.K.; Kim, H.J.; Werring, D.J.; Lee, J.S.; Choe, Y.S.; Park, S.; Lee, J.; Kim, K.W.; Kim, Y.; et al. Clinical significance of amyloid beta positivity in patients with probable cerebral amyloid angiopathy markers. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 1287–1298.
  85. Schmechel, D.E.; Saunders, A.M.; Strittmatter, W.J.; Crain, B.J.; Hulette, C.M.; Joo, S.H.; Pericak-Vance, M.A.; Goldgaber, D.; Roses, A.D. Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc. Natl. Acad. Sci. USA 1993, 90, 9649–9653.
  86. Kok, E.; Haikonen, S.; Luoto, T.; Huhtala, H.; Goebeler, S.; Haapasalo, H.; Karhunen, P.J. Apolipoprotein E-dependent accumulation of Alzheimer disease-related lesions begins in middle age. Ann. Neurol. 2009, 65, 650–657.
  87. Greenberg, S.M.; Vonsattel, J.-P.G.; Segal, A.Z.; Chiu, R.I.; Clatworthy, A.E.; Liao, A.; Hyman, B.T.; Rebeck, G.W. Association of apolipoprotein E ϵ2 and vasculopathy in cerebral amyloid angiopathy. Neurology 1998, 50, 961–965.
More