Open access peer-reviewed chapter

In Vivo Studies of Protein Misfolding and Neurodegeneration Induced by Metabolic Syndrome Relative to Chronic Cerebral Hypoperfusion: A Critical Review

Written By

María I. Herrera, Juan P. Luaces, Lucas D. Udovin, Nicolás Toro-Urrego, Matilde Otero-Losada and Francisco Capani

Submitted: 16 June 2019 Reviewed: 20 April 2020 Published: 26 November 2020

DOI: 10.5772/intechopen.92603

From the Edited Volume

Neuroprotection - New Approaches and Prospects

Edited by Matilde Otero-Losada, Francisco Capani and Santiago Perez Lloret

Chapter metrics overview

503 Chapter Downloads

View Full Metrics

Abstract

Metabolic syndrome (MetS) leads to microvascular dysfunction and chronic cerebral hypoperfusion (CCH) in an insidious way. Clinical evidence and several rodent models have contributed to determining the neurodegenerative effect of a sustained decrease in cerebral blood flow (CBF). Protein misfolding and aggregation derived from CCH might account for the establishment of vascular cognitive impairment and dementia (VCID) and Alzheimer’s disease (AD). However, the complex and multifactorial etiology of cerebrovascular disease demands the combination of experimental models in scientific research. In this sense, the present work aims at summarizing the differential available rodent paradigms for studying the establishment of cognitive decline resulting from protein misfolding induced by MetS in association with CCH. Revising experimental findings in the field will help further basic research on the pathophysiology of cerebrovascular disease and the future testing of protein-remodeling factors as neuroprotective agents for the prevention of cognitive impairment.

Keywords

  • metabolic syndrome (MetS)
  • chronic cerebral hypoperfusion (CCH)
  • protein misfolding
  • experimental models
  • cognitive impairment

1. Introduction

Metabolic syndrome (MetS) is the resulting condition of specific concurrent maladies, whose common pathogenic component is insulin resistance. Difficult to diagnose in clinical practice, there is consensus on its presence provided a cluster of risk factors be present, including abdominal obesity, hyperglycemia, hypertriglyceridemia, and hypertension [1, 2, 3]. Several murine models have contributed to the knowledge of this vascular risk factors’ constellation, which has been studied for over 80 years [4]. The experimental evidence shows that MetS silently, though relentlessly, leads to microvascular dysfunction and chronic cerebral hypoperfusion (CCH) [5]. Clinical findings, including the multivariate association between functional microvascular variables and laboratory-anthropometrical measurements [6], have reinforced the linkage of MetS with CCH [7], which leads to cognitive decline in late middle-aged adults [8]. As much as CCH might explain the considerable overlap between features of vascular cognitive impairment and dementia (VCID) and Alzheimer’s disease (AD), it might also underly as a common pathophysiological mechanism [9]. Experimental models of CCH have also contributed to exploring the interplay between hypoperfusion and amyloid β (Aβ) deposition, as it relates to AD [9]. Scientific evidence has underscored the importance of treating dementia comorbid disease conditions, including hypometabolism and diminished cerebral blood flow (CBF) [10]. An alternative target in neuroprotection is the regulation of the proteostasis network since protein aggregates link MetS-induced CCH and sporadic AD late-onset [11]. Therefore, the present work aims at revising different murine models of MetS and CCH, summarizing those experimental findings of relevance in the establishment of cerebrovascular disease. Plus, this overview intends to shed light on the usefulness of experimental models for the study of protein misfolding as a mechanism of neurodegeneration in CCH. Thirdly, this review attempts to discuss the requirement of combining MetS and CCH experimental models in order to resemble multifactorial conditions like VCID and AD and to test protein-remodeling factors as potential neuroprotective mechanisms for cognitive decline in the aging brain [12].

Advertisement

2. Experimental models of MetS and CCH: relevant findings to vascular dementia

Although MetS is a multifactorial and complex condition, several rat strains have been developed to assemble a profile of anomalies described in human subjects that exhibit cerebrovascular disease. Obese Zucker rats constitute the most representative rat strain to study this syndrome since animals present changes similar to those seen in patients [1]. This widely extended model of insulin resistance and obesity was discovered in 1961 by Lois Zucker. The mutation in the leptin receptor fa leads to noticeable obesity from the third week of life [13]. Leptin is synthesized by adipose tissue. This hormone acts in the brain on leptin receptors [14]. Elevated levels of leptin represent the molecular base of the characteristic phenotype of Zucker rats, which includes hyperphagia, deposition of energy in adipose tissue, dyslipidemia, mild glucose intolerance, hyperinsulinemia, and vascular changes [1, 15]. In contraposition to obese Zucker rats, the Wistar Ottawa Karlsburg W (WOKW) rat model is not induced by a single-gene mutation, resembling the context in which this pathology is developed in human subjects. However, these animals exhibit signs of MetS between 8 and 10 weeks of age, much later than Zucker rats [1].

Several murine models of MetS derive from the spontaneously hypertensive rat (SHR), which represents the of-choice experimental model of essential (or primary) hypertension. While the SHR rats show hypertriglyceridemia and abdominal obesity, corpulent SHR rats are preferable for reproducing MetS [1]. Different strains have been developed, including obese SHR or Koletsky rats, SHR/NIH-corpulent (SHR/N-cp) rats and its subline, the SHR/NDmc-corpulent rats, and stroke-prone-SHR fatty rats. The first strain, originally developed by Koletsky in 1970, shows premature vascular pathology mimicking human atherosclerosis [16]. The SHR/N corpulent model was established to reproduce obesity and non-insulin-dependent diabetes mellitus (NIDDM) [17]. Spontaneously hypertensive rats (SHR), an animal model of essential (or primary) hypertension, and SHR/NDmc-corpulent rats are also obese, presenting hyperphagia and metabolic alterations, while stroke-prone-SHR fatty rats are characterized by severe hypertension, which induces atherosclerosis and stroke. The spontaneously hypertensive/NIH-corpulent (SHR/N-cp) rat is a genetic model doomed to developing both non-insulin-dependent diabetes mellitus and hypertension.

Low-capacity runner (LCR) rats have been lately described, when cardiovascular risk factors were observed to emerge after artificial selection of low aerobic capacity [18]. These animals are selectively bred according to their performance in a running task. The LCR group is represented by rats capable of running short distances due to their low intrinsic aerobic capacity and bred with each other. After 11 generations, elevated blood pressure, insulin resistance, hyperinsulinemia, and endothelial dysfunction were registered in this strain [1]. Finally, from a translational perspective, both high-fat diet (HFD) and sweet carbonated beverage drinking represent two interesting rodent models of MetS evoking unhealthy dietary habits, increasing cardiovascular risk [19]. The former experimental paradigm reproduces impaired glucose tolerance (IGT) and type 2 diabetes. Rodents fed a HFD containing near 58% of total energy supply from fat develop obesity over the first week of life due to higher energy intake in combination with lower metabolic efficiency [20]. In the latter, 6-month ad libitum coke beverage drinking as the only liquid source results in hyperglycemia, hypertriglyceridemia, hypercholesterolemia, overweight, systolic hypertension, cardiac, renal alterations, and oxidative stress [2, 3, 21, 22, 23, 24]. Table 1 offers a translational overview of the abovementioned experimental models of MetS.

Name of the model Experimental induction of MetS Characteristic phenotype Translational advantages
Obese Zucker rats Mutation in leptin receptor fa causes obesity in rats from the 3rd week of life. Hyperphagia, energy deposition in adipose tissue, dyslipidemia, mild glucose intolerance, hyperinsulinemia, and vascular changes. Reproduces phenotypic changes resembling those in patients with MetS.
Wistar Ottawa Karlsburg W (WOKW) rats Derived from a Wistar rat outbred strain, WOKW rats first exhibit signs of MetS at 8–10 weeks of age. Obesity, moderate hypertension, dyslipidemia, hyperinsulinemia, and glucose intolerance. Resembles MetS in a polygenetic context, as in humans.
Spontaneously hypertensive rats (SHR) Rats bred with high blood pressure develop hypertension around 5–6 weeks of age. Hypertension, hypertriglyceridemia, and abdominal obesity. The phenotype varies according to the respective corpulent strain:
  1. Obese SHR or Koletsky rats

  2. SHR/NIH-corpulent (SHR/N-cp) rats

  3. SHR/NDmc-corpulent rats

  4. Stroke-prone-SHR fatty rats

The of-choice experimental model of essential (or primary) hypertension.
Corpulent SHR rats are preferable for reproducing MetS. Some strains resemble human-like atherosclerosis (a,d) or non-insulin-dependent (type II) diabetes mellitus (NIDDM) (b,c), respectively.
Low-capacity runner (LCR) rats Rats capable of running short distances due to their low intrinsic aerobic capacity are selectively bred with each other along various generations. Elevated blood pressure, insulin resistance, hyperinsulinemia, and endothelial dysfunction. Represents metabolic dysfunction associated with low aerobic capacity.
High-fat diet (HFD) Rodents fed a diet containing near 58% of total energy supply from fat become obese over the 1st. week of life due to higher energy intake. Obesity and low metabolic efficiency. Evokes impaired glucose tolerance (IGT) and type-2 diabetes due to unhealthy dietary habits.
Sweet carbonated beverage drinking 6-month ad libitum coke beverage drinking as the only liquid source causes metabolic dysfunction in rats. Hyperglycemia, hypertriglyceridemia, hypercholesterolemia, overweight, systolic hypertension, cardiorenal alterations, and oxidative stress. It mimics MetS derived from unhealthy dietary habits.

Table 1.

Experimental models of MetS: summary of phenotypic features from a translational perspective.

Disruption of CBF has been studied using focal or global ischemia. Focal ischemia models are used for resembling stroke pathophysiology and consist in the occlusion of a specific vessel, which reduces CBF by 70% due to restrictions in the vessel’s territory. This condition is generally induced by transient or permanent middle cerebral artery occlusion. Multiple infarcts can be reproduced via intra-arterial injection of emboli (heterogeneous localization) or by inducing spontaneous strokes (SHRSP). Higher reductions of CBF are developed in global ischemia models, which include transient common carotid artery occlusion (TCAO), three- and four-vessel occlusion, and cardiac arrest [9].

Since focal and global ischemia leads to severe reductions in CBF, alternative models have been developed to reproduce CCH, i.e., the subtle yet sustained decrease in CBF relevant to VCID. Early pathological events provoking VCID were studied through the ligation or occlusion of unilateral or bilateral common carotid arteries (two-vessel occlusion) [25]. Bilateral common carotid artery occlusion (BCCAO) was refined to resemble modest reductions in CBF. Bilateral common carotid artery stenosis (BCCAS) was developed to reduce flow to 50% of baseline [26]. However, flow largely recovered 1 month later, which was overcome by establishing a gradual stenosis model. Aneroid devices were used to absorb extracellular fluid and provoke the constriction of arteries, resulting in a slower and progressive onset of hypoperfusion. This experimental condition is known as gradual common carotid artery stenosis [27]. Consequently, murine models of CCH include a wide spectrum of disease severity, ranging from traditional occlusion mechanisms to gradual stenosis methods. Despite these variants, experimental models of CCH induce sustained and moderate blood flow reductions by 30–50%, in contraposition to ischemic models that reduce CBF in 70% acutely [9].

Although stenosis represents a better theoretical approach from a clinical perspective, it involves difficult techniques, rendering BCCAO the most commonly used model [28]. An alternative experimental model of CCH comprises the asymmetric common carotid artery surgery. Differential procedures are used for each common carotid artery (CCA), allowing interesting comparisons between both hemispheres. Gradual occlusion of the right artery lasts 1 month, while the left artery undergoes 50% stenosis by placing a micro-coil. Further investigation is necessary to assess CBF reductions at longer time points, discarding the complete occlusion of carotid arteries in the long term [9]. Table 2 summarizes the main features of the described models of CCH. For more details regarding experimental paradigms of CCH, including primate models, see [29].

Name of the model Experimental induction of CCH Characteristic phenotype Translational advantages
Bilateral common carotid artery occlusion (BCCAO) Both common carotid arteries are ligated. Cerebral blood flow (CBF) CBF rapidly decreases. Represents a widely used model of CCH, characterized by its feasibility.
Bilateral common carotid artery stenosis (BCCAS) Microcoils are placed on both CCAs. CBF decreases and gradually recovers. Mimics the clinical scenario of modest reductions in CBF.
Gradual common carotid artery stenosis (GCCAS) Aneroid devices are used to absorb extracellular fluid and provoke the constriction of arteries. CBF gradually decreases without recovery. Reproduces a progressive onset of hypoperfusion, slower than induced by BCCAS.
Asymmetric common carotid artery surgery (ACCAS) An aneroid constrictor is placed on the right common carotid artery (CCA), inducing a gradual occlusion for 1 month. The left CCA undergoes 50% stenosis by placing a micro-coil. CBF decreases to different extents between the right and left CCAs. Resembles differential reductions in CBF between both hemispheres.

Table 2.

Experimental models of CCH: Summary of phenotypic features from a translational perspective.

Recent evidence using the abovementioned experimental models of CCH has shown that disruption of CBF leads to vascular cognitive impairment (VCI). Because of BCCAS induction in mice, selective recognition alterations were encountered in the novel object recognition (NOR) test, together with damage to the perirhinal cortex [30]. When CBF was gradually reduced, progressive motor impairment and working memory decline were found in the rotarod and Y-maze tests, respectively. Loss of oligodendrocytes in the white matter might underlie these behavioral deficits, suggesting that the GCCAS model could closely replicate the clinical pathogenesis of hypoperfusive vascular dementia in humans [31]. After implanting an aneroid constrictor on the left CCA and provoking stenosis on the right CCA, mice exhibited sustained motor, learning, and memory dysfunction, inferred from the balance beam maze, a fear conditioning task, and the NOR test. Histopathological analysis showed neurodegeneration in the cerebral cortex, dorsal striatum, and hippocampus. These findings support the usefulness of the ACCAS experimental model for reproducing the effect of microvascular occlusions on cognitive impairment [32].

Advertisement

3. Experimental findings supporting protein misfolding as a neurodegenerative mechanism in CCH

The causative role of CCH in cognitive impairment and AD has been reported in several studies using the BCCAO model, an experimental paradigm of easy application [28]. Differential mechanisms have been proposed as a potential link between CCH and neurodegeneration, including synaptic dysfunction, oxidative stress, neuronal loss, white matter lesion, neuroinflammation, and protein misfolding [28]. The latter appears as a novel target for neuroprotection, according to cumulative evidence [11]. Figure 1 illustrates how CCH alters proteostasis network, leading to protein misfolding and neurodegeneration.

Figure 1.

Protein misfolding as a neurodegenerative mechanism and novel neuroprotective target in CCH. Chronic cerebral hypoperfusion impairs proteostasis network, inducing protein misfolding. Under cell stress, proteostasis network surveillance systems degrade proteins through different mechanisms. Depending on the nature of misfolded proteins, different systems are activated such as ubiquitin-proteasome system or macroautophagy. Protein aggregates, are recognized by molecular chaperones, ubiquitinated and delivered to the autophagosome via Beclin-1 complex. Neuroprotective agents, which target proteostasis network, emerge as promising treatments for cognitive impairment following CCH.

Degeneration of hippocampal neurons was attributed to proteostasis network destruction and protein aggregation induced by BCCAO [33]. This murine model has also led to a sustained increase in autophagy related-proteins Beclin-1, light chain 3B (LC3-B), and P62, suggesting a defensive reaction against protein misfolding. In this study, although CBF returned to baseline, cognitive failure was irreversible and attributed to Aβ aggregation in the hippocampus [34]. This brain region is characterized by its vulnerability to CCH [35] and its association with memory and learning dysfunction in AD [36]. Hippocampal degeneration has also been related to BCCAO-induced macroautophagy and endoplasmic reticulum (ER) stress, as it was inferred from the expression of light chain 3 II (LC3-II), Beclin 1, CCAAT/enhancer-binding protein, and C/EBP homologous protein [37].

Besides BCCAO, oxygen-glucose deprivation (OGD) provokes autophagy upregulation and apoptosis [35]. Recent evidence extended these findings, reporting CCH-induced high levels of LC3-II and Beclin-1 along with ultrastructural markers of apoptosis in CA1 neurons, including nuclear pycnosis, autophagosomes, and autolysosomes. ER fragmentation and spatial working memory impairment appeared as subcellular and functional correlates [38]. In addition to autophagy [34, 35, 38] and macroautophagy [37], ubiquitin-proteasome system (UPS) appears as another proteostatic pathway altered as a consequence of experimental CCH, which leads to CA1 degeneration. Long-term decrease peptidase activity and accumulation of ubiquitinated protein aggregates were observed after ligating the left vein and draining the transverse sinus and the bilateral external carotid arteries [39]. Previous studies from this laboratory had suggested the removal of misfolded proteins was impaired by UPS downregulation [40], and cognitive decline might be associated with long-term potentiation inhibition [41].

Along with aggregation of extracellular Aβ, intracellular phosphorylated tau protein deposition constitutes a hallmark of AD [42]. Tau hyperphosphorylation was observed as a result of unilateral common carotid artery occlusion (UCCAO), together with decreased post-translational tau O-GlcNAcylation by β-N-acetylglucosamine, dysregulation of synaptic proteins, and memory deficits [43]. According to previous findings, brain glucose metabolic dysfunction might downregulate tau O-GlcNAcylation mediated by tau hyperphosphorylation [44, 45, 46]. Recent studies have confirmed and extended this finding, suggesting CCH might exacerbate tau hyperphosphorylation in AD-rodents, either after UCCAO in mice [47] or BCCAO in rats [48]. Similarly, previous evidence prompted CCH might precipitate AD neuropathology since BCAS seemed to accelerate Aβ aggregation, the same process found in amyloid protein precursor (APP)-transgenic (APP-Tg) mice [49]. In fact, aberrant processing of APP has been reported after BCCAO [50].

Advertisement

4. Combining experimental models

Since MetS is associated with an increased risk of cerebral ischemia, recent investigations developed murine models of MetS combined with experimental paradigms of ischemia-reperfusion injury to study the impact of ischemia associated with MetS. Wistar rats fed a high-fat diet for 20 weeks were more susceptible to BCCAO-reperfusion than normal diet (ND)-fed animals, showing worsening in microvascular dysfunction and oxidative stress. These results show that MetS increases the vulnerability of the ischemic brain to damage, whereby BCCAO exacerbates cerebrovascular disease previously induced by HFD [51]. Similarly, BCCAO, followed by reperfusion, aggravated microvascular alterations in obese Zucker rats compared with lean Wistar controls. Lesions in the cortex and striatum were largely more pronounced in obese Zucker rats, suggesting MetS could increase the risk of adverse outcomes following a brain hypoperfusion-reperfusion event [52].

Novel findings support the hypothesis that the brain under obesity’s conditions is more vulnerable to ischemic injury. A brief episode of transient ischemia (TI) was provoked in obese gerbils, commonly known as desert rats. After 12 weeks of HFD, the rodents underwent a 2-min experimental TI. Hyperglycemia, cholesterolemia, and triglyceridemia observed in gerbils fed with a HFD were associated with a massive loss of pyramidal neurons in the hippocampal CA1 region 5 days after TI, indicating that a short-lived episode of TI could evoke neuronal damage along with pre-existing MetS. Increased levels of dihydroethidium, 4-hydroxynonenal, tumor necrosis factor-α, and interleukin-1β indicated brain injury was mediated by oxidative stress and neuroinflammation. On the contrary, ND gerbils did not exhibit neuronal death as a consequence of acute TI. In addition, these control animals could develop cerebral ischemic tolerance against a subsequent severer episode of TI [53]. Previous studies from the same laboratory have deepened the role of mammalian target of rapamycin (mTOR) in the pathogenesis of metabolic and neurological diseases and demonstrated that obesity and its related metabolic dysfunction might exacerbate the impact of TI in certain brain areas, including cerebral cortex, striatum, and hippocampus (CA1-3 regions) [54, 55, 56].

Conversely, cerebral ischemia itself might lead to glucose deregulation, a pathognomonic feature of MetS. Experimental evidence combining rodent models of ischemia and MetS shows that ischemic hippocampal neuronal death hampers glucose homeostasis, decreasing insulin secretion, which is later exacerbated by a HFD. In this study, gerbils were subjected to an 8-min BCCAO or a sham operation followed by either 11 or 40% fat diet for 7, 14, or 28 days. Although the initial occlusion provoked a 70% decrease in CA1 neurons, only HFD and longer ischemic periods resulted in larger hippocampal cell death. Similarly, glucose intolerance measured through the oral glucose tolerance test (OGTT) was overrated in gerbils fed a HFD as the ischemic periods became longer. During the OGTT, insulin levels were significantly lower in gerbils subjected to BCCAO than in sham-operated controls. In addition, insulin secretion decrease was elevated the most after 28 days of HFD in ischemic gerbils [57]. These interesting findings using a combination of rodent models suggest that ischemic damage is a risk factor for glucose homeostasis, which might be worsened by the experimental induction of MetS in a HFD paradigm.

Advertisement

5. Future directions

Scientific findings combining rodent models have shown that chronic MetS is associated with poor stroke outcomes following experimental cerebrovascular events [58] since HFD or the way of inducing MetS modulates ischemic mechanisms of brain damage [59]. Also, experimental CBF restriction seems to hamper glucose homeostasis, posing a risk factor for developing MetS. When artery occlusion models are followed by the experimental induction of metabolic dysfunction features, the resulting MetS might exacerbate previously ischemia-induced glucose deregulation [57]. Therefore, combining experimental models offers an interesting scientific paradigm for elucidating the complexity of pathophysiological mechanisms underlying chronic cerebrovascular disease. In terms of Zhao and Gong [28], differential clinical scenarios may coexist in chronic pathologies where risk factors rarely exist alone or may even exert a causative role in some patients while acting as a consequence in others. In this regard, experimental studies using the combined application of murine models could help to close the gap between rodent models and human disease [9].

However, this complex translational perspective is still necessary for studying the interaction between MetS and CCH inducing neurodegeneration. Although brain microvascular dysfunction has been confirmed in several murine models of MetS, including HFD [60, 61], Zucker [62], and SHR rats [63], whether MetS causes cognitive impairment due to a decrease in CBF has not been fully addressed yet [64]. In addition, since protein misfolding is a hallmark of neurodegenerative diseases [65], dissecting the exact role of MetS in association with CCH in protein aggregation represents a relevant challenge in the field. Detailed studies on the time-dependent proteostatic changes after experimental MetS and CCH will shed light on the roles and mechanisms of these clinical conditions in the establishment of VCID and AD. Furthermore, these studies will contribute to testing protein-remodeling factors [12] as putative neuroprotective agents for the prevention of cerebrovascular disease and cognitive decline.

References

  1. 1. de Artiñano AA, Castro MM. Experimental rat models to study the metabolic syndrome. British Journal of Nutrition. 2009;102(9):1246-1253. DOI: 10.1017/S0007114509990729
  2. 2. Otero-Losada M, González J, Müller A, Ottaviano G, Cao G, Azzato F, et al. Exercise ameliorates endocrine pancreas damage induced by chronic cola drinking in rats. PLoS One. 2016;11(5):e0155630. DOI: 10.1371/journal.pone.0155630
  3. 3. Otero-Losada M, Gómez Llambí H, Ottaviano G, Cao G, Müller A, Azzato F, et al. Cardiorenal involvement in metabolic syndrome induced by cola drinking in rats: Proinflammatory cytokines and impaired antioxidative protection. Mediators of Inflammation. 2016;2016:5613056. DOI: 10.1155/2016/5613056
  4. 4. Burton-Freeman B. Dietary fiber and energy regulation. The Journal of Nutrition. 2000;130(2):272S-275S. DOI: 10.1093/jn/130.2.272S
  5. 5. Aoqui C, Chmielewski S, Scherer E, Eißler R, Sollinger D, Heid I, et al. Microvascular dysfunction in the course of metabolic syndrome induced by high-fat diet. Cardiovascular Diabetology. 2014;13(1):31. DOI: 10.1186/1475-2840-13-31
  6. 6. Panazzolo DG, Sicuro FL, Clapauch R, Maranhão PA, Bouskela E, Kraemer-Aguiar LG. Obesity, metabolic syndrome, impaired fasting glucose, and microvascular dysfunction: A principal component analysis approach. BMC Cardiovascular Disorders. 2012;12(1):102. DOI: 10.1186/1471-2261-12-102
  7. 7. Czernichow S, Thomas D, Bruckert E. n-6 fatty acids and cardiovascular health: A review of the evidence for dietary intake recommendations. British Journal of Nutrition. 2010;104(6):788-796. DOI: 10.1017/S0007114510002096
  8. 8. Birdsill AC, Carlsson CM, Willette AA, Okonkwo OC, Johnson SC, Xu G, et al. Low cerebral blood flow is associated with lower memory function in metabolic syndrome. Obesity. 2013;21(7):1313-1320. DOI: 10.1002/oby.20170
  9. 9. Duncombe J, Kitamura A, Hase Y, Ihara M, Kalaria RN, Horsburgh K. 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. Clinical Science. 2017;131(19):2451-2468. DOI: 10.1042/CS20160727
  10. 10. Daulatzai MA. Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer’s disease. Journal of Neuroscience Research. 2017;95(4):943-972. DOI: 10.1002/jnr.23777
  11. 11. Herrera MI, Udovin LD, Toro-Urrego N, Kusnier CF, Luaces JP, Otero-Losada M, et al. Neuroprotection targeting protein misfolding on chronic cerebral hypoperfusion in the context of metabolic syndrome. Frontiers in Neuroscience. 2018;12(339):1-9. DOI: 10.3389/fnins.2018.00339
  12. 12. Jackrel ME, Shorter J. Protein-remodeling factors as potential therapeutics for neurodegenerative disease. Frontiers in Neuroscience. 2017;11:99. DOI: 10.3389/fnins.2017.00099
  13. 13. Phillips MS, Liu Q, Hammond HA, Dugan V, Hey PJ, Caskey CT, et al. Leptin receptor missense mutation in the fatty Zucker rat. Nature Genetics. 1996;13(1):18. DOI: 10.1038/ng0596-18
  14. 14. Ahima RS, Flier JS. Leptin. Annual Review of Physiology. 2000;62(1):413-437. DOI: 10.1146/annurev.physiol.62.1.413
  15. 15. Picó C, Sánchez J, Oliver P, Palou A. Leptin production by the stomach is up-regulated in obese (fa/fa) Zucker rats. Obesity Research. 2002;10(9):932-938. DOI: 10.1038/oby.2002.127
  16. 16. Kastin AJ, Pan W, Maness LM, Koletsky RJ, Ernsberger P. Decreased transport of leptin across the blood–brain barrier in rats lacking the short form of the leptin receptor. Peptides. 1999;20(12):1449-1453. DOI: 10.1016/s0196-9781(99)00156-4
  17. 17. Michaelis OE, Carswell N, Velasquez MT, Hansen CT. The role of obesity, hypertension and diet in diabetes and its complications in the spontaneous hypertensive/NIH-corpulent rat. Nutrition (Burbank, Los Angeles County, Calif.). 1989;5(1):56-59
  18. 18. Wisløff U, Najjar SM, Ellingsen Ø, Haram PM, Swoap S, Al-Share Q, et al. Cardiovascular risk factors emerge after artificial selection for low aerobic capacity. Science. 2005;307(5708):418-420. DOI: 10.1126/science.1108177
  19. 19. Abdel-Megeid FY, Abdelkarem HM, El-Fetouh AM. Unhealthy nutritional habits in university students are a risk factor for cardiovascular diseases. Saudi Medical Journal. 2011;32(6):621-627
  20. 20. Winzell MS, Ahrén B. The high-fat diet–fed mouse: A model for studying mechanisms and treatment of impaired glucose tolerance and type 2 diabetes. Diabetes. 2004;53(suppl 3):S215-S219. DOI: 10.2337/diabetes.53.suppl_3.S215
  21. 21. Otero-Losada M, Grana DR, Müller A, Ottaviano G, Ambrosio G, Milei J. Lipid profile and plasma antioxidant status in sweet carbonated beverage-induced metabolic syndrome in rat. International Journal of Cardiology. 2011;146(1):106-109. DOI: 10.1016/j.ijcard.2010.09.066
  22. 22. Otero-Losada M, Mc Loughlin S, Rodríguez-Granillo G, Müller A, Ottaviano G, Moriondo M, et al. Metabolic disturbances and worsening of atherosclerotic lesions in ApoE−/− mice after cola beverages drinking. Cardiovascular Diabetology. 2013;12(1):57. DOI: 10.1186/1475-2840-12-57
  23. 23. Otero-Losada M, Cao G, Mc Loughlin S, Rodríguez-Granillo G, Ottaviano G, Milei J. Rate of atherosclerosis progression in ApoE−/− mice long after discontinuation of cola beverage drinking. PLoS One. 2014;9(3):e89838. DOI: 10.1371/journal.pone.0089838
  24. 24. Otero-Losada M, Cao G, González J, Muller A, Ottaviano G, Lillig C, et al. Functional and morphological changes in endocrine pancreas following cola drink consumption in rats. PLoS One. 2015;10(3):e0118700. DOI: 10.1371/journal.pone.0118700
  25. 25. Farkas E, Luiten PG, Bari F. Permanent, bilateral common carotid artery occlusion in the rat: A model for chronic cerebral hypoperfusion-related neurodegenerative diseases. Brain Research Reviews. 2007;54(1):162-180. DOI: 10.1016/j.brainresrev.2007.01.003
  26. 26. Shibata M, Ohtani R, Ihara M, Tomimoto H. White matter lesions and glial activation in a novel mouse model of chronic cerebral hypoperfusion. Stroke. 2004;35(11):2598-2603. DOI: 10.1161/01.STR.0000143725.19053.60
  27. 27. Kitamura A, Fujita Y, Oishi N, Kalaria RN, Washida K, Maki T, et al. Selective white matter abnormalities in a novel rat model of vascular dementia. Neurobiology of Aging. 2012;33(5):1012.e25-1012.e35. DOI: 10.1016/j.neurobiolaging.2011.10.033
  28. 28. Zhao Y, Gong CX. From chronic cerebral hypoperfusion to Alzheimer-like brain pathology and neurodegeneration. Cellular and Molecular Neurobiology. 2015;35(1):101-110. DOI: 10.1007/s10571-014-0127-9
  29. 29. Washida K, Hattori Y, Ihara M. Animal models of chronic cerebral hypoperfusion: From mouse to primate. International Journal of Molecular Sciences. 2019;20(24):6176. DOI: 10.3390/ijms20246176
  30. 30. Patel A, Moalem A, Cheng H, Babadjouni RM, Patel K, Hodis DM, et al. Chronic cerebral hypoperfusion induced by bilateral carotid artery stenosis causes selective recognition impairment in adult mice. Neurological Research. 2017;39(10):910-917. DOI: 10.1080/01616412.2017.1355423
  31. 31. Hattori Y, Enmi JI, Iguchi S, Saito S, Yamamoto Y, Tsuji M, et al. Gradual carotid artery stenosis in mice closely replicates hypoperfusive vascular dementia in humans. Journal of the American Heart Association. 2016;5(2):e002757. DOI: 10.1161/JAHA.115.002757
  32. 32. Mehla J, Lacoursiere S, Stuart E, McDonald RJ, Mohajerani MH. Gradual cerebral hypoperfusion impairs fear conditioning and object recognition learning and memory in mice: Potential roles of neurodegeneration and cholinergic dysfunction. Journal of Alzheimer’s Disease. 2018;61(1):283-293. DOI: 10.3233/JAD-170635
  33. 33. Jian H, Yi-Fang W, Qi L, Xiao-Song H, Gui-Yun Z. Cerebral blood flow and metabolic changes in hippocampal regions of a modified rat model with chronic cerebral hypoperfusion. Acta Neurologica Belgica. 2013;113(3):313-317. DOI: 10.1007/s13760-012-0154-6
  34. 34. Zou W, Song Y, Li Y, Du Y, Zhang X, Fu J. The role of autophagy in the correlation between neuron damage and cognitive impairment in rat chronic cerebral hypoperfusion. Molecular Neurobiology. 2017;55(1):776-791. DOI: 10.1007/s12035-016-0351-z
  35. 35. Liu L, Li CJ, Lu Y, Zong XG, Luo C, Sun J, et al. Baclofen mediates neuroprotection on hippocampal CA1 pyramidal cells through the regulation of autophagy under chronic cerebral hypoperfusion. Scientific Reports. 2015;5:14474. DOI: 10.1038/srep14474
  36. 36. Ashraf MG, Greig HN, Khan TA, Hassan I, Tabrez S, Shakil S, et al. Protein misfolding and aggregation in Alzheimer’s disease and type 2 diabetes mellitus. CNS & Neurological Disorders Drug Targets. 2014;13(7):1280-1293. DOI: 10.2174/1871527313666140917095514
  37. 37. Jia Y, Jin W, Xiao Y, Dong Y, Wang T, Fan M, et al. Lipoxin A4 methyl ester alleviates vascular cognition impairment by regulating the expression of proteins related to autophagy and ER stress in the rat hippocampus. Cellular and Molecular Biology Letters. 2015;20(3):475-487. DOI: d10.1515/cmble-2015-0027
  38. 38. Wang D, Lin Q, Su S, Liu K, Wu Y, Hai J. URB597 improves cognitive impairment induced by chronic cerebral hypoperfusion by inhibiting mTOR-dependent autophagy. Neuroscience. 2017;344:293-304. DOI: 10.1016/j.neuroscience.2016.12.034
  39. 39. Hai J, Lin Q, Wu YF, Huang XS, Zhang GY, Wang F. Effects of N-stearoyl-L-tyrosine on the hippocampal ubiquitin-proteasome system in rats with chronic cerebral hypoperfusion. Neurological Research. 2013;35(7):734-743. DOI: 10.1179/1743132812Y.0000000154
  40. 40. Hai J, Lin Q, Su SH, Zhang L, Wan JF, Lu Y. Chronic cerebral hypoperfusion in rats causes proteasome dysfunction and aggregation of ubiquitinated proteins. Brain Research. 2011;1374:73-81. DOI: 10.1016/j.brainres.2010.12.030
  41. 41. Hai J, Wan JF, Lin Q, Wang F, Zhang L, Li H, et al. Cognitive dysfunction induced by chronic cerebral hypoperfusion in a rat model associated with arteriovenous malformations. Brain Research. 2009;1301:80-88. DOI: 10.1016/j.brainres.2009.09.001
  42. 42. Ozcan L, Tabas I. Role of endoplasmic reticulum stress in metabolic disease and other disorders. Annual Review of Medicine. 2012;63:317-328. DOI: 10.1146/annurev-med-043010-144749
  43. 43. Zhao Y, Gu JH, Dai CL, Liu Q, Iqbal K, Liu F, et al. Chronic cerebral hypoperfusion causes decrease of O-GlcNAcylation, hyperphosphorylation of tau and behavioral deficits in mice. Frontiers in Aging Neuroscience. 2014;6:10. DOI: 10.3389/fnagi.2014.00010
  44. 44. Liu F, Iqbal K, Grundke-Iqbal I, Hart GW, Gong CX. O-GlcNAcylation regulates phosphorylation of tau: A mechanism involved in Alzheimer’s disease. Proceedings of the National Academy of Sciences. 2004;101(29):10804-10809. DOI: 10.1073/pnas.0400348101
  45. 45. Liu F, Shi J, Tanimukai H, Gu J, Gu J, Grundke-Iqbal I, et al. Reduced O-GlcNAcylation links lower brain glucose metabolism and tau pathology in Alzheimer’s disease. Brain. 2009;132(7):1820-1832. DOI: 10.1093/brain/awp099
  46. 46. Liu Y, Liu F, Grundke-Iqbal I, Iqbal K, Gong CX. Brain glucose transporters, O-GlcNAcylation and phosphorylation of tau in diabetes and Alzheimer’s disease. Journal of Neurochemistry. 2009;111(1):242-249. DOI: 10.1111/j.1471-4159.2009.06320.x
  47. 47. Qiu L, Ng G, Tan EK, Liao P, Kandiah N, Zeng L. Chronic cerebral hypoperfusion enhances tau hyperphosphorylation and reduces autophagy in Alzheimer’s disease mice. Scientific Reports. 2016;6:23964. DOI: 10.1038/srep23964
  48. 48. Li JF, Wang Z, Sun QJ, Du YF. Expression of tau protein in rats with cognitive dysfunction induced by cerebral hypoperfusion. International Journal of Clinical and Experimental Medicine. 2015;8(10):19682-19688
  49. 49. Kitaguchi H, Tomimoto H, Ihara M, Shibata M, Uemura K, Kalaria RN, et al. Chronic cerebral hypoperfusion accelerates amyloid β deposition in APPSwInd transgenic mice. Brain Research. 2009;1294:202-210. DOI: 10.1016/j.brainres.2009.07.078
  50. 50. Bennett SA, Pappas BA, Stevens WD, Davidson CM, Fortin T, Chen J. Cleavage of amyloid precursor protein elicited by chronic cerebral hypoperfusion. Neurobiolology of Aging. 2000;21(2):207-214. DOI: 10.1016/S0197-4580(00)00131-7
  51. 51. Obadia N, Lessa MA, Daliry A, Silvares RR, Gomes F, Tibiriçá E, et al. Cerebral microvascular dysfunction in metabolic syndrome is exacerbated by ischemia–reperfusion injury. BMC Neuroscience. 2017;18(1):67. DOI: 10.1186/s12868-017-0384-x
  52. 52. Lapi D, Mastantuono T, Sapio D, Paterni M, Colantuoni A. Pial microvascular responses induced by transient bilateral common carotid artery occlusion in Zucker rats. Clinical Hemorheology and Microcirculation. 2013;54(4):415-429. DOI: 10.3233/CH-131763
  53. 53. Park JH, Ahn JH, Song M, Kim H, Park CW, Park YE, et al. A 2-min transient ischemia confers cerebral ischemic tolerance in non-obese gerbils, but results in neuronal death in obese gerbils by increasing abnormal mTOR activation-mediated oxidative stress and Neuroinflammation. Cell. 2019;8(10):1126. DOI: 10.3390/cells8101126
  54. 54. Ahn JH, Shin MC, Kim DW, Kim H, Song M, Lee TK, et al. Antioxidant properties of Fucoidan alleviate acceleration and exacerbation of hippocampal neuronal death following transient global cerebral ischemia in high-fat diet-induced obese gerbils. International Journal of Molecular Sciences. 2019;20(3):554. DOI: 10.3390/ijms20030554
  55. 55. Song M, Ahn JH, Kim H, Kim DW, Lee TK, Lee JC, et al. Chronic high-fat diet-induced obesity in gerbils increases pro-inflammatory cytokines and mTOR activation, and elicits neuronal death in the striatum following brief transient ischemia. Neurochemistry International. 2018;121:75-85. DOI: 10.1016/j.neuint.2018.09.009
  56. 56. Yan BC, Park JH, Ahn JH, Kim IH, Lee JC, Yoo KY, et al. Effects of high-fat diet on neuronal damage, gliosis, inflammatory process and oxidative stress in the hippocampus induced by transient cerebral ischemia. Neurochemical Research. 2014;39(12):2465-2478. DOI: 10.1007/s11064-014-1450-8
  57. 57. Park S, Kim DS, Kang S, Kwon DY. Ischemic hippocampal cell death induces glucose dysregulation by attenuating glucose-stimulated insulin secretion which is exacerbated by a high fat diet. Life Sciences. 2011;88(17-18):766-773. DOI: 10.1016/j.lfs.2011.02.021
  58. 58. Chantler PD, Shrader CD, Tabone LE, d’Audiffret AC, Huseynova K, Brooks SD, et al. Cerebral cortical microvascular rarefaction in metabolic syndrome is dependent on insulin resistance and loss of nitric oxide bioavailability. Microcirculation. 2015;22(6):435-445. DOI: 10.1111/micc.12209
  59. 59. Moon SM, Choi GM, Yoo DY, Jung HY, Yim HS, Kim DW, et al. Differential effects of pioglitazone in the hippocampal CA1 region following transient forebrain ischemia in low-and high-fat diet-fed gerbils. Neurochemical Research. 2015;40(5):1063-1073. DOI: 10.1007/s11064-015-1568-3
  60. 60. Estato V, Nascimento A, Antunes B, Gomes F, Coelho L, Rangel R, et al. Cerebral microvascular dysfunction and inflammation are improved by centrally acting antihypertensive drugs in metabolic syndrome. Metabolic Syndrome and Related Disorders. 2017;15(1):26-35. DOI: 10.1089/met.2016.0085
  61. 61. Nascimento AR, Machado MV, Gomes F, Vieira AB, Gonçalves-de-Albuquerque CF, Lessa MA, et al. Central sympathetic modulation reverses microvascular alterations in a rat model of high-fat diet-induced metabolic syndrome. Microcirculation. 2016;23(4):320-329. DOI: 10.1111/micc.12280
  62. 62. Brooks SD, DeVallance E, d’Audiffret AC, Frisbee SJ, Tabone LE, Shrader CD, et al. Metabolic syndrome impairs reactivity and wall mechanics of cerebral resistance arteries in obese Zucker rats. American Journal of Physiology. Heart and Circulatory Physiology. 2015;309(11):H1846-H1859. DOI: 10.1152/ajpheart.00691.2015
  63. 63. Estato V, Obadia N, Carvalho-Tavares J, Freitas FS, Reis P, Neto HCF, et al. Blockade of the renin–angiotensin system improves cerebral microcirculatory perfusion in diabetic hypertensive rats. Microvascular Research. 2013;87:41-49. DOI: 10.1016/j.mvr.2013.02.007
  64. 64. Zuloaga KL, Johnson LA, Roese NE, Marzulla T, Zhang W, Nie X, et al. High fat diet-induced diabetes in mice exacerbates cognitive deficit due to chronic hypoperfusion. Journal of Cerebral Blood Flow & Metabolism. 2016;36(7):1257-1270. DOI: 10.1177/0271678X15616400
  65. 65. Sweeney P, Park H, Baumann M, Dunlop J, Frydman J, Kopito R, et al. Protein misfolding in neurodegenerative diseases: Implications and strategies. Translational Neurodegeneration. 2017;6(1):6. DOI: 10.1186/s40035-017-0077-5

Written By

María I. Herrera, Juan P. Luaces, Lucas D. Udovin, Nicolás Toro-Urrego, Matilde Otero-Losada and Francisco Capani

Submitted: 16 June 2019 Reviewed: 20 April 2020 Published: 26 November 2020