Next Article in Journal
Zileuton Alleviates Radiation-Induced Cutaneous Ulcers via Inhibition of Senescence-Associated Secretory Phenotype in Rodents
Previous Article in Journal
TM4SF5-Mediated Regulation of Hepatocyte Transporters during Metabolic Liver Diseases
Previous Article in Special Issue
Glucose Stimulates Glial Cell Line-Derived Neurotrophic Factor Gene Expression in Microglia through a GLUT5-Independent Mechanism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Relationship between Brain Metabolic Disorders and Cognitive Impairment: LDL Receptor Defect

1
Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea
2
Soonchunhyang Institute of Medi-Bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(15), 8384; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23158384
Submission received: 14 June 2022 / Revised: 26 July 2022 / Accepted: 27 July 2022 / Published: 29 July 2022

Abstract

:
The low-density-lipoprotein receptor (LDLr) removes low-density lipoprotein (LDL), an endovascular transporter that carries cholesterol from the bloodstream to peripheral tissues. The maintenance of cholesterol content in the brain, which is important to protect brain function, is affected by LDLr. LDLr co-localizes with the insulin receptor and complements the internalization of LDL. In LDLr deficiency, LDL blood levels and insulin resistance increase, leading to abnormal cholesterol control and cognitive deficits in atherosclerosis. Defects in brain cholesterol metabolism lead to neuroinflammation and blood–brain-barrier (BBB) degradation. Moreover, interactions between endoplasmic reticulum stress (ER stress) and mitochondria are induced by ox-LDL accumulation, apolipoprotein E (ApoE) regulates the levels of amyloid beta (Aβ) in the brain, and hypoxia is induced by apoptosis induced by the LDLr defect. This review summarizes the association between neurodegenerative brain disease and typical cognitive deficits.

1. Introduction

Low-density-lipoprotein receptor (LDLr) is involved in the regulation of blood cholesterol. LDLr internalizes cholesterol-containing LDL ligands, and insulin receptor (IR) maintains glycemic homeostasis. Although the exact mechanism is not completely clear, diabetes is associated with dyslipidemia. Diabetes is also characterized by abnormalities ranging from IR to absolute deficiency of insulin (type 1 diabetes) or abnormalities in the receptor protein itself post-transcription/translation/translation (type 2 diabetes). The LDL-clearing activity of LDLr depends on the interaction between insulin and IR [1].
Insulin has been reported to stimulate LDLr expression, an IR-dependent signaling event [2]. The inactivation of IR and LDLr prevents the removal of extracellular LDL and promotes hyperproteinemia through intravascular LDL deposition. In insulin-deficient type 1 diabetes and insulin-resistant type 2 diabetes, IR inactivation leads to LDLr inactivation, which increases the severity of atherosclerotic complications due to the inability to remove vascular LDL.
Cholesterol in the brain builds up the membrane surfaces of large numbers of axon dendrites and synapses, including post-synaptic spines and pre-synaptic vesicles [3,4,5]. The brain comprises neurons and glial cells, which build up a large amount of membrane and occupy a high area and volume; thus, the cholesterol requirement is very high. Cholesterol metabolism is important in neurons and glial cells, which should cooperate for brain development and function, and LDLr, which interacts with IR, plays an important role in cholesterol synthesis and turnover regulation [6]. Cholesterol depletion in neurons impairs synaptic vesicle exocytosis, nerve activity, and neurotransmission, leading to dendritic spine and synaptic degeneration [7,8,9]. Furthermore, defects in cholesterol metabolism lead to structural and functional central nervous system (CNS) diseases such as Huntington’s, Alzheimer’s, and Parkinson’s diseases [10,11,12].
The hippocampus of an LDLr−/− rodent model fed a high-cholesterol diet showed a higher neuroinflammatory response and impaired blood–brain-barrier (BBB) transport [13,14,15]. This affected the proliferation of hippocampal progenitors. Additionally, the neuroinflammatory process increases the production of ROS, which is detrimental to neurons [16]. Mitochondria are prone to oxidative damage, and inflammation can further contribute to mitochondrial dysfunction [17] and GSH-dependent antioxidant system damage. Several studies have reported an association between high dietary exposure to fat or cholesterol and oxidative stress in the rat and rat brain [18,19].
It is necessary to prove the correlation between diseases such as hypercholesterolemia, which are caused due to LDLr defects, with respect to brain metabolic physiology. Therefore, this review aims to provide a detailed overview of LDLr-defect-mediated metabolism disorders.

2. Cholesterol Regulation of LDLr

LDLr is a cell membrane glycoprotein that LDL, a cholesterol transporter, binds and internalizes. LDLr is a key receptor for maintaining cholesterol homeostasis by removing LDL through endocytosis, and is essential for lipoprotein and lipid metabolism. [20,21,22,23,24,25].
When cholesterol accumulates or decreases, the endoplasmic reticulum (ER) detects the level of membrane cholesterol and activates the cholesterol regulatory system (Figure 1), the sterol regulatory element-binding protein (SREBP) pathway, to maintain cholesterol homeostasis. When the intracellular cholesterol level is low, SREBP forms a complex with the polytopic membrane protein SREBP cleavage-activating protein (Scap) in the form of vesicles coated with coat protein complex II (COPII) in the ER, and the vesicles are then transported to the Golgi. Upon transporting the Scap/SREBP complex, SREBP is proteolytically degraded into an active fragment, which activates genes involved in cholesterol synthesis and absorption [26,27]. SREBP2 is activated by a reduction in intracellular cholesterol and induction of genes such as proprotein convertase subtilisin/kexin type 9 (PCSK9) and LDLr, leading to the endocytosis of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) LDL [27,28,29]. On the other hand, when the cholesterol level is high, the Scap/SREBP complex binds to Insig-1 or Insig-2, another polytopic membrane protein, and the coating of the Scap/SREBP complex is blocked by COPII and maintained in the ER. By preventing SREBP from moving to the Golgi [30,31,32], the transcription of target gene decreases cholesterol synthesis and absorption [28].
LDLr relies on ARH, a low-density-lipoprotein receptor adapter protein, for LDL internalization. ARH is mediated via S-nitrosylation by nitric oxide, and LDL is absorbed into the LDLr. In ARH−/− cells, LDLr activity is inhibited due to ARH loss via LDLr endocytosis failure by the induction of LDLr redistribution into the plasma membrane [33,34,35,36,37]. PCSK9 can degrade LDLr internalized in lysosomes and protect cells from excessive LDL uptake and cholesterol accumulation [38]. HMGCR activates the acetyl-CoA pathway, producing cholesterol as the final product [39].

3. Interaction of LDLr and IR

LDLr, a transmembrane glycoprotein, is involved in regulating blood cholesterol by binding and internalizing LDL containing cholesterol [40,41,42,43]. LDLr removes cholesterol-containing LDL particles [41,44], and IR maintains glycemic homeostasis [45,46,47]. On the other hand, although the exact mechanism is not completely clear, diabetes is associated with dyslipidemia [48]. Diabetes mellitus is characterized by metabolic abnormalities due to insufficient insulin production due to loss of beta cells (type 1 diabetes) or abnormalities in the insulin receptor protein itself (type 2 diabetes). Insulin is known to regulate several biological activities [49,50,51], resulting in increased glucose transport and the maintenance of adequate blood glucose levels [52].
LDLr and IR co-localization in organelles has been observed via electron microscopy. Increased insulin levels increase the LDL uptake of HepG2 cells via LDLr due to the disruption of the LDLr–IR co-localized complex; they further promote the internalization of extracellular LDL particles by directly regulating insulin-mediated LDLr function [53,54]. We have demonstrated the co-localization of LDLr and IR by reducing the expression of LDLr and IR-related proteins via LDLr-specific siRNA treatment [1].
In LDL exposure, autophagosome formation is suppressed via PI3K/Akt/mTOR activation, a key autophagy regulator in HUVECs, similar to the insulin pathway, and LC3 and p62 expression in the lipidized form of the autophagosome decreases. After LDL treatment, LDLr and IR expression increases in the cytosol rather than the membrane, indicating endocytosis from the cell membrane to cytosol, opposite to GLUT1’s translocation from the cell cytosol to membrane [55,56].
The impairment of IR function is associated with abnormal glucose metabolism leading to impaired glucose tolerance or diabetes, a condition of obesity and insulin resistance [57]. Insulin resistance is also associated with intravascular LDL accumulation due to poor clearance by functionally impaired LDLr. Insulin resistance results in decreased expression levels of insulin and IGF receptors, which promote neurite growth, synapse formation, and neuronal survival in the brains of Alzheimer’s patients [58]. The intraventricular injection of streptozotocin, which induces diabetes in rats, or the depletion of neuronal IR results in cognitive decline [59,60,61,62,63].

4. Cholesterol Metabolism in the Brain

Cholesterol is a structural component of the membrane that acts as a buffer for changes in the fluidity of cell lipid membranes and is involved in membrane-intrinsic proteins and cell signal transduction [64,65]. Despite these important functions, high cholesterol levels can be toxic to cells. An overload of total cellular cholesterol in the plasma membrane triggers its migration to the ER, resulting in the depletion of calcium stores, leading to cell death [66,67]. Additionally, a loss of membrane fluidity due to increased cholesterol levels can lead to dysfunction of the integral membrane protein and damage to the membrane domain, resulting in the disruption of signaling events [68].
Consequently, sterol-sensing proteins that regulate cholesterol homeostasis, sterol synthesis and degradation by regulatory mechanisms, and LDLr are also involved [69]. The brain is the most cholesterol-rich organ in the human body and contains 25% cholesterol [70,71]. Cholesterol homeostasis may influence neuroinflammatory expression for PCSK9 and neuronal receptors. PCSK9, which can regulate the cholesterol receptor LDLr and apolipoprotein E (ApoE), maintains a certain concentration of cholesterol in the human cerebrospinal fluid under normal conditions [72,73]. Under increased BBB permeability in disease states, PCSK9 crosses the BBB and induces LDLr degradation in the brain [74,75].
High levels of LDL-C have been observed in stroke patients with mutations in LDLr-related genes. Sequencing has been performed in stroke patients with LDLr, apolipoprotein B (ApoB), and PCSK9 gene targets, and the familial hypercholesterolemia pathogenic gene may indicate atherosclerotic phenotypes such as increased carotid intima–media thickness and ischemic attacks [76,77,78,79].

5. BBB Breakdown

The BBB is formed and maintained by cerebral capillaries, pericytes surrounding the capillaries, and endothelial cells surrounding astrocyte ends that surround these two layers. The BBB, composed of adherens junctions (AJs) and tight junctions (TJs), selectively regulates the transport of molecules and cells in and out of the brain, thereby regulating the brain microenvironment [80,81]. It is composed of TJs comprising transmembrane proteins such as occludin and claudins and AJs comprising VE-cadherin, which plays an important role in maintaining brain endothelial junctions. The BBB maintains the homeostasis of neurovascular units, including blood vessels and nerve cells [82].
Many studies have demonstrated the potential for BBB disruption to alter brain regions and dissociation states, allowing neurotoxic plasma components, blood cells, and pathogens to enter the brain and cause neuroinflammation [83,84]. LDLr−/− mice exposed to a high-cholesterol diet and accompanied by a decreased gene expression of claudin-5 and occludin and increased BBB permeability to stimulate neuroinflammation and cognition are more susceptible to the disorder, and BBB disruption is associated with brain changes due to hypercholesterolemia [75]. In LDLr−/− mice fed a high-cholesterol diet, aquaporin-4 (AQP-4) expression increases [85,86], occludin and claudin-5 gene expression decreases, BBB permeability increases, and GFAP-derived neuroinflammation increases in the hippocampus [75].

6. Neuroinflammation

LDLr is important for regulating LDL homeostasis. LDL is oxidized, producing oxLDL. Although LDL-C levels decrease in the sera of patients with rheumatoid arthritis, a chronic inflammatory disease, high levels of oxLDL [87] produce cytokines such as TNF-a and IL-6 [88,89]. After cerebral I/R injury, the downregulation of LDLr expression in neurons, astrocytes, and oligodendrocytes [90] leads to neurological deficits, infarction, and edema in the CNS. Additionally, LDLr KO activates pattern recognition receptors (PRRs) present in innate immune cells with damage-associated molecular patterns (DAMPs) generated in damaged cells or tissues (Figure 2) [91], and in the middle cerebral artery occlusion (MCAO) model under ischemic conditions. Additionally, LDL accumulation was proven to be inflammatory by inducing TLR activation via pathogen-associated molecular patterns (PAMPs) [92,93], and oxLDL, acting as a DAMP and PAMP, forms the NLRP3 inflammasome complex [94,95]. The complex induces gasdermin D (GSDMD)-mediated lytic apoptosis (pyroptosis) by increasing the levels of active caspase-1. Pyroptosis, a programmed cell death process, is mediated by a pore formed by the binding of GSDMD N-terminal fragments cleaved by active caspase-1 to the plasma membrane, inducing the release of inflammatory cytokines such as IL-1β and IL-18, leading to neuronal pyroptosis [96,97,98,99,100]. Additionally, interleukin-10 (IL-10), an anti-inflammatory cytokine, inhibits the activity of inflammatory cytokines by inhibiting TLR induction [93].
The blockade of NLRP3 delays neuronal pyroptosis in LDLr−/− mice and cultured LDLr−/− neurons after experimental stroke [91]. Oxidative stress and mitochondrial DNA (mtDNA) damage have been observed in atherosclerotic plaques in LDLr−/− mice, and the number of atherosclerotic plaques increase due to an additional deficiency of Ogg1 promoting NLRP3 inflammasome activation in LDLr−/− mice. The mitochondria and mtDNA, which are sites for reactive oxygen species (ROS) production, are vulnerable to oxidative stress due to a lack of protective histones. Ogg1, a glycosylase responsible for the cleavage of 7,8-dihydro-8-oxo-2’-deoxyguanosine, a byproduct of ROS, and AMPK, an upstream regulator, are targets of miR-33, a proatherogenic microRNA. As a result, the levels of Ogg1 and AMPK decrease in atherosclerotic plaques [101].
LDLr overexpression is associated with inflammation relief. Lipopolysaccharides (LPS), which are made up of lipids and polysaccharides, can cause inflammation. They have the highest binding affinity for high-density lipoprotein (HDL), and HDL-binding LPS is redistributed to LDL and VLDL [102]. Pathogen lipids such as LPS are integrated and transduced into lipoprotein particles such as HDL, LDL, and VLDL, triggering an immune-inflammatory response through Toll-like receptors (TLRs). TLRs are mammalian PRRs that recognize structural pathogen-associated molecular patterns shared by microorganisms in innate immunity. The inhibition of PCSK9, which promotes the degradation of LDLr lysosomes, increases the ability of LDLr to remove pathogenic lipids, thereby reducing the inflammatory response [103,104].

7. Interaction between ER Stress and Mitochondria

LDLr deficiency can lead to cognitive impairment due to interactions between ER stress and mitochondria. In particular, the brain is vulnerable to oxidative damage and apoptosis induction by abnormal calcium and ATP levels due to its high energy metabolism rate, high oxygen consumption, and high ratio of polyunsaturated fatty acids [105,106,107,108,109,110]. Oxidative stress is attracting attention as a cause of several neurodegenerative diseases [111,112,113].
LDL is oxidized to oxLDL, which is taken up by lectin-like oxidized low-density-lipoprotein-1 receptors (Figure 3). OxLDL increases lectin-like oxidized LDL receptor-1 (LOX-1) expression in macrophages, leading to macrophage migration and foam cell differentiation, leading to deposition into endothelial cells [114,115]. OxLDL uptake induces Ca2+ overload, which further induces mitochondrial dysfunction, leading to cytochrome c release, the apoptosis of endothelial cells, and the suppression of antioxidant activity, generating ROS that can activate NF-κB [116,117,118,119,120,121,122].
Abnormally folded proteins in the ER lumen are increased in atherosclerosis resulting from cholesterol accumulation. The three major ER-stress sensor proteins of the UPR that remove these abnormally folded proteins are inositol-requiring enzyme 1 (IRE1), activating transcription factor 6 (ATF6) and protein kinase RNA-like ER kinase (PERK). In the steady state, the three sensor proteins remain inactive due to their binding to the ER chaperone, glucose response protein-78 (BiP/GRP78). Stress releases BiP from the ER sensor and induces the phosphorylation and dimerization of IRE1 and PERK in cancers, whereby active PERK promotes the phosphorylation of eukaryotic initiation factor 2α (eIF2α) and inhibits protein translation by the activation of activating transcription factor 4 (ATF4) [123,124], and X-box binding protein 1 (XBP1) expression is achieved through ATF6 activation [125]. However, prolonged cellular stress induces CHOP expression, and a CHOP-mediated imbalance in the Bcl-2 family activates proapoptotic proteins in the mitochondrial membrane, inducing the release of cytochrome c, leading to subsequent mitochondrion-dependent apoptosis [126,127].
In pathological conditions such as ischemia and reperfusion, the depolarization of the mitochondrial membrane potential increases, causing Ca2+ overload in the outer membrane and Ca2+ conduction through the calcium uniporter (MCU), which is locally present in the inner mitochondrial membrane. Increased Ca2+ is produced due to a bidirectional interaction with ROS [128,129,130,131]. The mitochondrial outer membrane channel is formed by the opening of the mitochondrial permeability transition pore (mPTP), which induces mitochondrial membrane permeability and the insertion of proapoptotic BH3-domain-containing proteins such as Bcl-2-associated X protein (Bax). In addition, due to the depletion of cardiolipin, a lipid constituting the inner membrane of mitochondria, cytochrome c is released from the inner membrane of mitochondria into the cytoplasm [132,133]. Cytochrome c forms an apoptosome and induces apoptosis by forming a complex with apoptosis-protease activating factor 1, which is required for the proteolysis of caspase-9 and caspase-3 [134,135]. In the mitochondria, ROS are formed as a byproduct of oxidative phosphorylation that induces ATP production. Imbalances in ROS, ATP, and Ca2+ that appear in mitochondrial dysfunction increase the expression of antioxidant enzymes, such as superoxide dismutase (SOD), glutathione (GSH), and glutathione peroxidase (GPx), and the Bcl-2-mediated apoptosis mechanism [136]. Furthermore, oxidative stress also affects endothelial cells.
Hypercholesterolemia-induced LDLr−/− mice have shown a decreased activity of mitochondrial complex I and II in the cerebral cortex; decreased GSH; an approximately 40% increase in complexes formed via thiobarbituric acid’s reaction with malondialdehyde (MDA), a product of lipid peroxidation; and enzymes related to peroxide removal. An imbalance in phosphorus GPx/glutathione reductase activity results in mitochondrial dysfunction and oxidative stress, leading to cognitive impairment [137,138]. In 14-month-old LDLr−/− mice, antioxidant imbalance and glutathione metabolism increase due to brain oxidative stress because of lipid peroxidation, and the acetylcholinesterase activity, which degrades the neurotransmitter acetylcholine, also increases [139]. The memory deficit observed in LDLr−/− mice is not related to Aβ-level changes in the prefrontal cortex and hippocampus; however, Bcl-2 expression and caspase-3 activation decrease, while Bax expression increases.

8. ApoE

Low-density-lipoprotein receptor (LDLR) and apolipoprotein E (ApoE) are responsible for the transport of cholesterol-rich lipoproteins (Figure 4). Deficiencies in LDLR and ApoE are associated with increased plasma total cholesterol and, consequently, a higher risk of hypercholesterolemia, atherosclerosis, and coronary artery disease [140,141,142]. ApoE is mainly produced by astrocytes and is a protein that plays a role in lipid transport in the CNS [143]. Unlike the LDLR-binding molecule ApoB100, small fat-soluble molecules such as ApoE can cross the BBB-forming membrane and affect BBB stability [144]. ApoE was found to co-localize with amyloid plaque [145,146]. ApoE exists in three isoforms: ApoE2, ApoE3, and ApoE4. ApoE4 induces brain damage as traumatic brain injury (TBI) [147], Alzheimer’s disease [148], and conditions leading to impaired cognition [149] do.
LDLR is an important receptor for ApoE in the central nervous system. [10.1016/j.nbd.2004.01.015] (accessed on 26 July 2022) Amyloid beta (Aβ), known to cause Alzheimer’s disease, is hypothesized to accumulate in the brain as plaques and is regulated by receptors for ApoE [150,151,152]. LDLr is expressed in astrocytes and induces the uptake of ApoE and Aβ [153]. It prevents Aβ deposition by reducing ApoE levels in LDLr-overexpressing mice [154]. LDLr is associated with the major central nervous system ApoE receptor regulating amyloid deposition in a distinct mouse model of β-amyloidosis. The acute intraventricular injection of aggregated Aβ(1–40) peptides was used to show increased susceptibility to Aβ-induced neurotoxicity, with intrahippocampal oxidative stress, neuroinflammation, nerve membrane damage, memory deficits, and increased blood–brain-barrier permeability [155].
ApoE also affects the immune response, and in mice, ApoE KO increased the expression of Toll-like receptor 4 (TLR4) and LOX-1, suggesting the formation of foam cells and promoting the onset of arteriosclerosis [156]. Additionally, ApoE promotes phagocytosis by binding to triggering receptor expressed on myeloid cells 2 (TREM2) in microglia [157]. LDLr−/− mice exhibit reduced numbers of synaptophysin-immunoreactive presynaptic boutons in hippocampus CA1 compared to LDLr+/+ mice, resulting in hippocampus-dependent memory function impairment [0].

9. Conclusions

Our analysis revealed that LDLr defects might lead to IR interactions, BBB breakdown, neuroinflammatory responses, interactions between ER stress and mitochondria, and hypoxia. There are various cells in the brain that make up a complex cellular network. Each cell exhibits a lipid membrane structure as an essential element for cell maintenance and signal transduction, and lipids are then delivered by LDL, which is regulated by LDLr.
However, studies on LDLr defects have mainly been conducted in the liver, which is the main organ for lipid synthesis; thus, there is a lack of studies on changes in hormones such as cortisol, insulin, and leptin secreted into the brain signaling system and the mechanism of LDLr regulation specifically for brain cells.
In conclusion, this review summarizes the relationship between LDLr defects and brain metabolism. Further detailed studies are required to elucidate the mechanisms involved in LDLr.

Author Contributions

Conceptualization, D.-Y.H., D.-H.L., M.-R.L. and J.-S.O.; methodology, D.-Y.H. and D.-H.L.; software, D.-Y.H., D.-H.L., J.-Y.L., E.-C.L., S.-W.P. and J.-S.O.; validation, D.-Y.H., D.-H.L., M.-R.L. and J.-S.O.; formal analysis, D.-Y.H. and D.-H.L.; investigation, D.-Y.H., D.-H.L. and J.-S.O.; resources, M.-R.L. and J.-S.O.; data curation, D.-Y.H., D.-H.L. and J.-S.O.; writing—original draft preparation, D.-Y.H., D.-H.L., M.-R.L. and J.-S.O.; writing—review and editing, D.-Y.H., D.-H.L., M.-R.L. and J.-S.O.; visualization, D.-Y.H. and D.-H.L.; supervision, M.-R.L. and J.-S.O.; project administration, M.-R.L. and J.-S.O.; funding acquisition, M.-R.L. and J.-S.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a grant from the Soonchunhyang University Fund, by the Bio & Medical Technology Development Program of the National Research Foundation funded by the Korean government (NRF-2019M3E5D1A02069061 & 2020R1F1A1066362).

Institutional Review Board Statement

This research was performed in compliance with the Institutional Animal Care and Use Committee of Soon Chun Hyang University (IACUC No. SCH 20-0065).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Ramakrishnan, G.; Arjuman, A.; Suneja, S.; Das, C.; Chandra, N.C. The association between insulin and low-density lipoprotein receptors. Diabetes Vasc. Dis. Res. 2012, 9, 196–204. [Google Scholar]
  2. Wade, D.P.; Knight, B.L.; Soutar, A.K. Hormonal regulation of low-density lipoprotein (LDL) receptor activity in human hepatoma Hep G2 cells: Insulin increases LDL receptor activity and diminishes its suppression by exogenous LDL. Eur. J. Biochem. 1988, 174, 213–218. [Google Scholar]
  3. Goritz, C.; Mauch, D.H.; Pfrieger, F.W. Multiple mechanisms mediate cholesterol-induced synaptogenesis in a CNS neuron. Mol. Cell. Neurosci. 2005, 29, 190–201. [Google Scholar]
  4. Pfenninger, K.H. Plasma membrane expansion: A neuron’s Herculean task. Nat. Rev. Neurosci. 2009, 10, 251–261. [Google Scholar]
  5. Takamori, S.; Holt, M.; Stenius, K.; Lemke, E.A.; Grønborg, M.; Riedel, D.; Urlaub, H.; Schenck, S.; Brügger, B.; Ringler, P.; et al. Molecular anatomy of a trafficking organelle. Cell 2006, 127, 831–846. [Google Scholar]
  6. Pfrieger, F.W.; Ungerer, N. Cholesterol metabolism in neurons and astrocytes. Prog. Lipid Res. 2011, 50, 357–371. [Google Scholar]
  7. Linetti, A.; Fratangeli, A.; Taverna, E.; Valnegri, P.; Francolini, M.; Cappello, V.; Matteoli, M.; Passafaro, M.; Rosa, P. Cholesterol reduction impairs exocytosis of synaptic vesicles. J. Cell Sci. 2010, 123, 595–605. [Google Scholar]
  8. Liu, Q.; Trotter, J.; Zhang, J.; Peters, M.M.; Cheng, H.; Bao, J.; Han, X.; Weeber, E.J.; Bu, G. Neuronal LRP1 knockout in adult mice leads to impaired brain lipid metabolism and progressive, age-dependent synapse loss and neurodegeneration. J. Neurosci. 2010, 30, 17068–17078. [Google Scholar]
  9. Liu, Q.; Zerbinatti, C.V.; Zhang, J.; Hoe, H.-S.; Wang, B.; Cole, S.L.; Herz, J.; Muglia, L.; Bu, G. Amyloid precursor protein regulates brain apolipoprotein E and cholesterol metabolism through lipoprotein receptor LRP1. Neuron 2007, 56, 66–78. [Google Scholar]
  10. Block, R.C.; Dorsey, E.R.; Beck, C.A.; Brenna, J.T.; Shoulson, I. Altered cholesterol and fatty acid metabolism in Huntington disease. J. Clin. Lipidol. 2010, 4, 17–23. [Google Scholar]
  11. Di Paolo, G.; Kim, T.-W. Linking lipids to Alzheimer’s disease: Cholesterol and beyond. Nat. Rev. Neurosci. 2011, 12, 284–296. [Google Scholar]
  12. Wang, Q.; Yan, J.; Chen, X.; Li, J.; Yang, Y.; Weng, J.; Deng, C.; Yenari, M.A. Statins: Multiple neuroprotective mechanisms in neurodegenerative diseases. Exp. Neurol. 2011, 230, 27–34. [Google Scholar]
  13. Thirumangalakudi, L.; Prakasam, A.; Zhang, R.; Bimonte-Nelson, H.; Sambamurti, K.; Kindy, M.S.; Bhat, N.R. High cholesterol-induced neuroinflammation and amyloid precursor protein processing correlate with loss of working memory in mice. J. Neurochem. 2008, 106, 475–485. [Google Scholar]
  14. Moreira, E.L.G.; de Oliveira, J.; Engel, D.F.; Walz, R.; de Bem, A.F.; Farina, M.; Prediger, R.D.S. Hypercholesterolemia induces short-term spatial memory impairments in mice: Up-regulation of acetylcholinesterase activity as an early and causal event? J. Neural Transm. 2014, 121, 415–426. [Google Scholar]
  15. Rutkowsky, J.M.; Lee, L.L.; Puchowicz, M.; Golub, M.S.; Befroy, D.E.; Wilson, D.W.; Anderson, S.; Cline, G.; Bini, J.; Borkowski, K.; et al. Reduced cognitive function, increased blood-brain-barrier transport and inflammatory responses, and altered brain metabolites in LDLr−/−and C57BL/6 mice fed a western diet. PLoS ONE 2018, 13, e0191909. [Google Scholar]
  16. Witte, M.E.; Geurts, J.J.; de Vries, H.E.; van der Valk, P.; van Horssen, J. Mitochondrial dysfunction: A potential link between neuroinflammation and neurodegeneration? Mitochondrion 2010, 10, 411–418. [Google Scholar]
  17. Yadav, R.; Weng, H.-R. EZH2 regulates spinal neuroinflammation in rats with neuropathic pain. Neuroscience 2017, 349, 106–117. [Google Scholar]
  18. Crisby, M.; Rahman, S.; Sylven, C.; Winblad, B.; Schultzberg, M. Effects of high cholesterol diet on gliosis in apolipoprotein E knockout mice: Implications for Alzheimer’s disease and stroke. Neurosci. Lett. 2004, 369, 87–92. [Google Scholar]
  19. Montilla, P.; Espejo, I.; Muñoz, M.C.; Bujalance, I.; Muñoz-Castañeda, J.R.; Tunez, I. Protective effect of red wine on oxidative stress and antioxidant enzyme activities in the brain and kidney induced by feeding high cholesterol in rats. Clin. Nutr. 2006, 25, 146–153. [Google Scholar]
  20. Goldstein, J.L.; Brown, M.S. Binding and degradation of low density lipoproteins by cultured human fibroblasts. Comparison of cells from a normal subject and from a patient with homozygous familial hypercholesterolemia. J. Biol. Chem. 1974, 249, 5153–5162. [Google Scholar]
  21. Goldstein, J.L.; Brown, M.S.; Stone, N.J. Genetics of the LDL receptor: Evidence that the mutations affecting binding and internalization are allelic. Cell 1977, 12, 629–641. [Google Scholar]
  22. Goldstein, J.L.; Dana, S.E.; Brown, M.S. Esterification of low density lipoprotein cholesterol in human fibroblasts and its absence in homozygous familial hypercholesterolemia. Proc. Natl. Acad. Sci. USA 1974, 71, 4288–4292. [Google Scholar]
  23. Goldstein, J.L.; Brown, M.S. Familial hypercholesterolemia: Identification of a defect in the regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity associated with overproduction of cholesterol. Proc. Natl. Acad. Sci. USA 1973, 70, 2804–2808. [Google Scholar]
  24. Brown, M.S.; Goldstein, J.L. Familial hypercholesterolemia: Defective binding of lipoproteins to cultured fibroblasts associated with impaired regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity. Proc. Natl. Acad. Sci. USA 1974, 71, 788–792. [Google Scholar]
  25. Brown, M.S.; Goldstein, J.L. Expression of the familial hypercholesterolemia gene in heterozygotes: Mechanism for a dominant disorder in man. Science 1974, 185, 61–63. [Google Scholar]
  26. Brown, M.S.; Goldstein, J.L. A proteolytic pathway that controls the cholesterol content of membranes, cells, and blood. Proc. Natl. Acad. Sci. USA 1999, 96, 11041–11048. [Google Scholar]
  27. Horton, J.D.; Shah, N.A.; Warrington, J.A.; Anderson, N.N.; Park, S.W.; Brown, M.S.; Goldstein, J.L. Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. Proc. Natl. Acad. Sci. USA 2003, 100, 12027–12032. [Google Scholar]
  28. Radhakrishnan, A.; Goldstein, J.L.; McDonald, J.G.; Brown, M.S. Switch-like control of SREBP-2 transport triggered by small changes in ER cholesterol: A delicate balance. Cell Metab. 2008, 8, 512–521. [Google Scholar]
  29. Yang, H.X.; Zhang, M.; Long, S.Y.; Tuo, Q.H.; Tian, Y.; Chen, J.X.; Zhang, C.P.; Liao, D.F. Cholesterol in LDL receptor recycling and degradation. Clin. Chim. Acta 2020, 500, 81–86. [Google Scholar]
  30. Sun, L.P.; Li, L.; Goldstein, J.L.; Brown, M.S. Insig required for sterol-mediated inhibition of Scap/SREBP binding to COPII proteins in vitro. J. Biol. Chem. 2005, 280, 26483–26490. [Google Scholar]
  31. Yang, T.; Espenshade, P.J.; Wright, M.E.; Yabe, D.; Gong, Y.; Aebersold, R.; Goldstein, J.L.; Brown, M.S. Crucial step in cholesterol homeostasis: Sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell 2002, 110, 489–500. [Google Scholar]
  32. Yabe, D.; Brown, M.S.; Goldstein, J.L. Insig-2, a second endoplasmic reticulum protein that binds SCAP and blocks export of sterol regulatory element-binding proteins. Proc. Natl. Acad. Sci. USA 2002, 99, 12753–12758. [Google Scholar]
  33. Garcia, C.K.; Wilund, K.; Arca, M.; Zuliani, G.; Fellin, R.; Maioli, M.; Calandra, S.; Bertolini, S.; Cossu, F.; Grishin, N.; et al. Autosomal recessive hypercholesterolemia caused by mutations in a putative LDL receptor adaptor protein. Science 2001, 292, 1394–1398. [Google Scholar]
  34. He, G.; Gupta, S.; Yi, M.; Michaely, P.; Hobbs, H.H.; Cohen, J.C. ARH is a modular adaptor protein that interacts with the LDL receptor, clathrin, and AP-2. J. Biol. Chem. 2002, 277, 44044–44049. [Google Scholar]
  35. Michaely, P.; Li, W.P.; Anderson, R.G.; Cohen, J.C.; Hobbs, H.H. The modular adaptor protein ARH is required for low density lipoprotein (LDL) binding and internalization but not for LDL receptor clustering in coated pits. J. Biol. Chem. 2004, 279, 34023–34031. [Google Scholar]
  36. Zhao, Z.; Pompey, S.; Dong, H.; Weng, J.; Garuti, R.; Michaely, P. S-nitrosylation of ARH is required for LDL uptake by the LDL receptor. J. Lipid Res. 2013, 54, 1550–1559. [Google Scholar]
  37. Tao, W.; Moore, R.; Meng, Y.; Smith, E.R.; Xu, X.X. Endocytic adaptors Arh and Dab2 control homeostasis of circulatory cholesterol. J. Lipid Res. 2016, 57, 809–817. [Google Scholar]
  38. Park, S.W.; Moon, Y.A.; Horton, J.D. Post-transcriptional regulation of low density lipoprotein receptor protein by proprotein convertase subtilisin/kexin type 9a in mouse liver. J. Biol. Chem. 2004, 279, 50630–50638. [Google Scholar]
  39. Zhang, J.; Liu, Q. Cholesterol metabolism and homeostasis in the brain. Protein Cell 2015, 6, 254–264. [Google Scholar]
  40. Goldstein, J.L.; Brown, M.S. The low-density lipoprotein pathway and its relation to atherosclerosis. Annu. Rev. Biochem. 1977, 46, 897–930. [Google Scholar]
  41. Brown, M.S.; Goldstein, J.L. A receptor-mediated pathway for cholesterol homeostasis. Science 1986, 232, 34–47. [Google Scholar]
  42. Jeon, H.; Blacklow, S.C. Structure and physiologic function of the low-density lipoprotein receptor. Annu. Rev. Biochem. 2005, 74, 535–562. [Google Scholar]
  43. Go, G.W.; Mani, A. Low-density lipoprotein receptor (LDLR) family orchestrates cholesterol homeostasis. Yale J. Biol. Med. 2012, 85, 19–28. [Google Scholar]
  44. Anderson, R.G.; Brown, M.S.; Goldstein, J.L. Role of the coated endocytic vesicle in the uptake of receptor-bound low density lipoprotein in human fibroblasts. Cell 1977, 10, 351–364. [Google Scholar]
  45. De Meyts, P. Insulin and its receptor: Structure, function and evolution. Bioessays 2004, 26, 1351–1362. [Google Scholar]
  46. De Meyts, P. The insulin receptor: A prototype for dimeric, allosteric membrane receptors? Trends Biochem. Sci. 2008, 33, 376–384. [Google Scholar]
  47. Youngren, J.F. Regulation of insulin receptor function. Cell Mol. Life Sci. 2007, 64, 873–891. [Google Scholar]
  48. Steiner, G. The dyslipoproteinemias of diabetes. Atherosclerosis 1994, 110, S27–S33. [Google Scholar]
  49. Ilonen, J.; Lempainen, J.; Veijola, R. The heterogeneous pathogenesis of type 1 diabetes mellitus. Nat. Rev. Endocrinol. 2019, 15, 635–650. [Google Scholar]
  50. Kahn, S.E.; Cooper, M.E.; del Prato, S. Pathophysiology and treatment of type 2 diabetes: Perspectives on the past, present, and future. Lancet 2014, 383, 1068–1083. [Google Scholar]
  51. Ozougwu, J.; Obimba, K.; Belonwu, C.; Unakalamba, C. The pathogenesis and pathophysiology of type 1 and type 2 diabetes mellitus. J. Physiol. Pathophysiol. 2013, 4, 46–57. [Google Scholar]
  52. Guerre-Millo, M.; Lavau, M.; Horne, J.S.; Wardzala, L.J. Proposed mechanism for increased insulin-mediated glucose transport in adipose cells from young, obese Zucker rats. Large intracellular pool of glucose transporters. J. Biol. Chem. 1985, 260, 2197–2201. [Google Scholar]
  53. Ill, C.; Lepine, J.; Gospodarowicz, D. Permissive effect of insulin on the adenosine 3′,5′-monophosphate-dependent up-regulation of low density lipoprotein receptors and the stimulation of steroid release in bovine adrenal cortical cells. Endocrinology 1984, 114, 767–775. [Google Scholar]
  54. Chandra, N.C. A comprehensive account of insulin and LDL receptor activity over the years: A highlight on their signaling and functional role. J. Biochem. Mol. Toxicol. 2021, 35, e22840. [Google Scholar]
  55. Zhu, L.; Wu, G.; Yang, X.; Jia, X.; Li, J.; Bai, X.; Li, W.; Zhao, Y.; Li, Y.; Cheng, W.; et al. Low density lipoprotein mimics insulin action on autophagy and glucose uptake in endothelial cells. Sci. Rep. 2019, 9, 3020. [Google Scholar]
  56. Tang, F.; Yang, T.L. MicroRNA-126 alleviates endothelial cells injury in atherosclerosis by restoring autophagic flux via inhibiting of PI3K/Akt/mTOR pathway. Biochem. Biophys. Res. Commun. 2018, 495, 1482–1489. [Google Scholar]
  57. Obici, S.; Feng, Z.; Karkanias, G.; Baskin, D.G.; Rossetti, L. Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nat. Neurosci. 2002, 5, 566–572. [Google Scholar]
  58. Liu, T.; Li, Y.; Yang, B.; Wang, H.; Lu, C.; Chang, A.K.; Huang, X.; Zhang, X.; Lu, Z.; Lu, X.; et al. Suppression of neuronal cholesterol biosynthesis impairs brain functions through insulin-like growth factor I-Akt signaling. Int. J. Biol. Sci. 2021, 17, 3702–3716. [Google Scholar]
  59. Blokland, A.; Jolles, J. Spatial learning deficit and reduced hippocampal ChAT activity in rats after an ICV injection of streptozotocin. Pharmacol. Biochem. Behav. 1993, 44, 491–494. [Google Scholar]
  60. Kinoshita, J.; Fagan, A.; Ewbank, D.; Marlatt, M.; Heyn, P.; de la Monte, S.; Lombardo, N.E. Alzheimer Research Forum live discussion: Insulin resistance: A common axis linking Alzheimer’s, depression, and metabolism? J. Alzheimers Dis. 2006, 9, 89–93. [Google Scholar]
  61. Reddy, P.H.; Mani, G.; Park, B.S.; Jacques, J.; Murdoch, G.; Whetsell, W., Jr.; Kaye, J.; Manczak, M. Differential loss of synaptic proteins in Alzheimer’s disease: Implications for synaptic dysfunction. J. Alzheimers Dis. 2005, 7, 103–117. [Google Scholar]
  62. de la Monte, S.M.; Tong, M.; Lester-Coll, N.; Plater, M., Jr.; Wands, J.R. Therapeutic rescue of neurodegeneration in experimental type 3 diabetes: Relevance to Alzheimer’s disease. J. Alzheimers Dis. 2006, 10, 89–109. [Google Scholar]
  63. Lester-Coll, N.; Rivera, E.J.; Soscia, S.J.; Doiron, K.; Wands, J.R.; de la Monte, S.M. Intracerebral streptozotocin model of type 3 diabetes: Relevance to sporadic Alzheimer’s disease. J. Alzheimers Dis. 2006, 9, 13–33. [Google Scholar]
  64. Simons, K.; Sampaio, J.L. Membrane organization and lipid rafts. Cold Spring Harb. Perspect. Biol. 2011, 3, a004697. [Google Scholar]
  65. Subczynski, W.K.; Pasenkiewicz-Gierula, M.; Widomska, J.; Mainali, L.; Raguz, M. High Cholesterol/Low Cholesterol: Effects in Biological Membranes: A Review. Cell Biochem. Biophys. 2017, 7, 369–385. [Google Scholar]
  66. Gliozzi, M.; Musolino, V.; Bosco, F.; Scicchitano, M.; Scarano, F.; Nucera, S.; Zito, M.C.; Ruga, S.; Carresi, C.; Macrì, R.; et al. Cholesterol homeostasis: Researching a dialogue between the brain and peripheral tissues. Pharmacol. Res. 2021, 163, 105215. [Google Scholar]
  67. Feng, B.; Yao, P.M.; Li, Y.; Devlin, C.M.; Zhang, D.; Harding, H.P.; Sweeney, M.; Rong, J.X.; Kuriakose, G.; Fisher, E.A.; et al. The endoplasmic reticulum is the site of cholesterol-induced cytotoxicity in macrophages. Nat. Cell Biol. 2003, 5, 781–792. [Google Scholar]
  68. Tabas, I. Consequences of cellular cholesterol accumulation: Basic concepts and physiological implications. J. Clin. Investig. 2002, 110, 905–911. [Google Scholar]
  69. Howe, V.; Sharpe, L.J.; Alexopoulos, S.J.; Kunze, S.V.; Chua, N.K.; Li, D.; Brown, A.J. Cholesterol homeostasis: How do cells sense sterol excess? Chem. Phys. Lipids 2016, 199, 170–178. [Google Scholar]
  70. Dietschy, J.M. Central nervous system: Cholesterol turnover, brain development and neurodegeneration. Biol. Chem. 2009, 390, 287–293. [Google Scholar]
  71. Dietschy, J.M.; Turley, S.D. Thematic review series: Brain Lipids. Cholesterol metabolism in the central nervous system during early development and in the mature animal. J. Lipid Res. 2004, 45, 1375–1397. [Google Scholar]
  72. Chen, Y.Q.; Troutt, J.S.; Konrad, R.J. PCSK9 is present in human cerebrospinal fluid and is maintained at remarkably constant concentrations throughout the course of the day. Lipids 2014, 49, 445–455. [Google Scholar]
  73. Zimetti, F.; Caffarra, P.; Ronda, N.; Favari, E.; Adorni, M.P.; Zanotti, I.; Bernini, F.; Barocco, F.; Spallazzi, M.; Galimberti, D.; et al. Increased PCSK9 Cerebrospinal Fluid Concentrations in Alzheimer’s Disease. J. Alzheimers Dis. 2017, 55, 315–320. [Google Scholar]
  74. Adorni, M.P.; Ruscica, M.; Ferri, N.; Bernini, F.; Zimetti, F. Proprotein Convertase Subtilisin/Kexin Type 9, Brain Cholesterol Homeostasis and Potential Implication for Alzheimer’s Disease. Front Aging Neurosci. 2019, 11, 120. [Google Scholar]
  75. de Oliveira, J.; Engel, D.F.; de Paula, G.C.; Santos, D.B.D.; Lopes, J.B.; Farina, M.; Moreira, E.L.G.; de Bem, A.F. High Cholesterol Diet Exacerbates Blood-Brain Barrier Disruption in LDLr−/− Mice: Impact on Cognitive Function. J. Alzheimers Dis. 2020, 78, 97–115. [Google Scholar]
  76. Tung, H.; Lin, H.J.; Chen, P.L.; Lu, T.J.; Jhan, P.P.; Chen, J.P.; Chen, Y.M.; Wu, C.C.; Lin, Y.Y.; Hsiao, T.-H. Characterization of familial hypercholesterolemia in Taiwanese ischemic stroke patients. Aging 2021, 13, 19339–19351. [Google Scholar]
  77. Khera, A.V.; Won, H.H.; Peloso, G.M.; Lawson, K.S.; Bartz, T.M.; Deng, X.; van Leeuwen, E.M.; Natarajan, P.; Emdin, C.A.; Bick, A.G.; et al. Diagnostic Yield and Clinical Utility of Sequencing Familial Hypercholesterolemia Genes in Patients With Severe Hypercholesterolemia. J. Am. Coll Cardiol. 2016, 67, 2578–2589. [Google Scholar]
  78. Mahdieh, N.; Heshmatzad, K.; Rabbani, B. A systematic review of LDLR, PCSK9, and APOB variants in Asia. Atherosclerosis 2020, 305, 50–57. [Google Scholar]
  79. Meshkov, A.; Ershova, A.; Kiseleva, A.; Zotova, E.; Sotnikova, E.; Petukhova, A.; Zharikova, A.; Malyshev, P.; Rozhkova, T.; Limonova, A.; et al. The LDLR, APOB, and PCSK9 Variants of Index Patients with Familial Hypercholesterolemia in Russia. Genes 2021, 12, 66. [Google Scholar]
  80. Wolburg, H.; Lippoldt, A. Tight junctions of the blood-brain barrier: Development, composition and regulation. Vascul Pharmacol. 2002, 38, 323–337. [Google Scholar]
  81. Li, W.; Chen, Z.; Chin, I.; Chen, Z.; Dai, H. The Role of VE-cadherin in Blood-brain Barrier Integrity Under Central Nervous System Pathological Conditions. Curr. Neuropharmacol. 2018, 16, 1375–1384. [Google Scholar]
  82. Abbott, N.J.; Rönnbäck, L.; Hansson, E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci. 2006, 7, 41–53. [Google Scholar]
  83. Tucsek, Z.; Toth, P.; Tarantini, S.; Sosnowska, D.; Gautam, T.; Warrington, J.P.; Giles, C.B.; Wren, J.D.; Koller, A.; Ballabh, P.; et al. Aging exacerbates obesity-induced cerebromicrovascular rarefaction, neurovascular uncoupling, and cognitive decline in mice. J. Gerontol. Biol. Sci. Med. Sci. 2014, 69, 1339–1352. [Google Scholar]
  84. Deng, J.; Zhao, F.; Yu, X.; Zhao, Y.; Li, D.; Shi, H.; Sun, Y. Expression of aquaporin 4 and breakdown of the blood-brain barrier after hypoglycemia-induced brain edema in rats. PLoS ONE 2014, 9, e107022. [Google Scholar]
  85. Mader, S.; Brimberg, L. Aquaporin-4 Water Channel in the Brain and Its Implication for Health and Disease. Cells 2019, 8, 90. [Google Scholar]
  86. Karpouzas, G.A.; Ormseth, S.R.; Ronda, N.; Hernandez, E.; Budoff, M.J. Lipoprotein oxidation may underlie the paradoxical association of low cholesterol with coronary atherosclerotic risk in rheumatoid arthritis. J. Autoimmun. 2022, 129, 102815. [Google Scholar]
  87. Saad, A.F.; Virella, G.; Chassereau, C.; Boackle, R.J.; Lopes-Virella, M.F. OxLDL immune complexes activate complement and induce cytokine production by MonoMac 6 cells and human macrophages. J. Lipid Res. 2006, 47, 1975–1983. [Google Scholar]
  88. Morris, G.; Berk, M.; Walder, K.; O’Neil, A.; Maes, M.; Puri, B.K. The lipid paradox in neuroprogressive disorders: Causes and consequences. Neurosci. Biobehav. Rev. 2021, 128, 35–57. [Google Scholar]
  89. Saher, G.; Stumpf, S.K. Cholesterol in myelin biogenesis and hypomyelinating disorders. Biochim. Biophys. Acta 2015, 1851, 1083–1094. [Google Scholar]
  90. Sun, R.; Peng, M.; Xu, P.; Huang, F.; Xie, Y.; Li, J.; Hong, Y.; Guo, H.; Liu, Q.; Zhu, W. Low-density lipoprotein receptor (LDLR) regulates NLRP3-mediated neuronal pyroptosis following cerebral ischemia/reperfusion injury. J. Neuroinflamm. 2020, 17, 330. [Google Scholar]
  91. Lee, D.H.; Lee, J.Y.; Hong, D.Y.; Lee, E.C.; Park, S.W.; Lee, M.R.; Oh, J.S. Neuroinflammation in Post-Traumatic Stress Disorder. Biomedicines 2022, 10, 953. [Google Scholar]
  92. Bzowska, M.; Nogieć, A.; Skrzeczyńska-Moncznik, J.; Mickowska, B.; Guzik, K.; Pryjma, J. Oxidized LDLs inhibit TLR-induced IL-10 production by monocytes: A new aspect of pathogen-accelerated atherosclerosis. Inflammation 2012, 35, 1567–1584. [Google Scholar]
  93. Imai, Y.; Kuba, K.; Neely, G.G.; Yaghubian-Malhami, R.; Perkmann, T.; van Loo, G.; Ermolaeva, M.; Veldhuizen, R.; Leung, Y.H.; Wang, H.; et al. Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Cell 2008, 133, 235–249. [Google Scholar]
  94. Jo, E.K.; Kim, J.K.; Shin, D.M.; Sasakawa, C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell Mol. Immunol. 2016, 13, 148–159. [Google Scholar]
  95. Barrington, J.; Lemarchand, E.; Allan, S.M. A brain in flame; do inflammasomes and pyroptosis influence stroke pathology? Brain Pathol. 2017, 27, 205–212. [Google Scholar]
  96. Malik, A.; Kanneganti, T.D. Inflammasome activation and assembly at a glance. J. Cell Sci. 2017, 130, 3955–3963. [Google Scholar]
  97. Zhang, D.; Qian, J.; Zhang, P.; Li, H.; Shen, H.; Li, X.; Chen, G. Gasdermin D serves as a key executioner of pyroptosis in experimental cerebral ischemia and reperfusion model both in vivo and in vitro. J. Neurosci. Res. 2019, 97, 645–660. [Google Scholar]
  98. Wang, Q.; Wu, J.; Zeng, Y.; Chen, K.; Wang, C.; Yang, S.; Sun, N.; Chen, H.; Duan, K.; Zeng, G. Pyroptosis: A pro-inflammatory type of cell death in cardiovascular disease. Clin. Chim. Acta 2020, 510, 62–72. [Google Scholar]
  99. Opoku, E.; Traughber, C.A.; Zhang, D.; Iacano, A.J.; Khan, M.; Han, J.; Smith, J.D.; Gulshan, K. Gasdermin D Mediates Inflammation-Induced Defects in Reverse Cholesterol Transport and Promotes Atherosclerosis. Front Cell Dev. Biol. 2021, 9, 715211. [Google Scholar]
  100. Tumurkhuu, G.; Shimada, K.; Dagvadorj, J.; Crother, T.R.; Zhang, W.; Luthringer, D.; Gottlieb, R.A.; Chen, S.; Arditi, M. Ogg1-Dependent DNA Repair Regulates NLRP3 Inflammasome and Prevents Atherosclerosis. Circ. Res. 2016, 119, e76–e90. [Google Scholar]
  101. Levels, J.H.; Marquart, J.A.; Abraham, P.R.; van den Ende, A.E.; Molhuizen, H.O.; van Deventer, S.J.; Meijers, J.C. Lipopolysaccharide is transferred from high-density to low-density lipoproteins by lipopolysaccharide-binding protein and phospholipid transfer protein. Infect Immun. 2005, 73, 2321–2326. [Google Scholar]
  102. Walley, K.R.; Thain, K.R.; Russell, J.A.; Reilly, M.P.; Meyer, N.J.; Ferguson, J.F.; Christie, J.D.; Nakada, T.A.; Fjell, C.D.; Thair, S.A.; et al. PCSK9 is a critical regulator of the innate immune response and septic shock outcome. Sci. Transl. Med. 2014, 6, 258ra143. [Google Scholar]
  103. Tang, Z.H.; Li, T.H.; Peng, J.; Zheng, J.; Li, T.T.; Liu, L.S.; Jiang, Z.S.; Zheng, X.L. PCSK9: A novel inflammation modulator in atherosclerosis? J. Cell Physiol. 2019, 234, 2345–2355. [Google Scholar]
  104. Chen, H.; Chan, D.C. Critical dependence of neurons on mitochondrial dynamics. Curr. Opin. Cell Biol. 2006, 18, 453–459. [Google Scholar]
  105. Sacks, D.; Baxter, B.; Campbell, B.C.V.; Carpenter, J.S.; Cognard, C.; Dippel, D.; Eesa, M.; Fischer, U.; Hausegger, K.; Hirsch, J.A.; et al. Multisociety Consensus Quality Improvement Revised Consensus Statement for Endovascular Therapy of Acute Ischemic Stroke. Int. J. Stroke. 2018, 13, 612–632. [Google Scholar]
  106. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar]
  107. Kizhakkedath, P.; John, A.; Al-Sawafi, B.K.; Al-Gazali, L.; Ali, B.R. Endoplasmic reticulum quality control of LDLR variants associated with familial hypercholesterolemia. FEBS Open Bio. 2019, 9, 1994–2005. [Google Scholar]
  108. Sørensen, S.; Ranheim, T.; Bakken, K.S.; Leren, T.P.; Kulseth, M.A. Retention of mutant low density lipoprotein receptor in endoplasmic reticulum (ER) leads to ER stress. J. Biol. Chem. 2006, 281, 468–476. [Google Scholar]
  109. Oommen, D.; Kizhakkedath, P.; Jawabri, A.A.; Varghese, D.S.; Ali, B.R. Proteostasis Regulation in the Endoplasmic Reticulum: An Emerging Theme in the Molecular Pathology and Therapeutic Management of Familial Hypercholesterolemia. Front Genet. 2020, 11, 570355. [Google Scholar]
  110. El-Bachá, R.S.; De-Lima-Filho, J.L.; Guedes, R.C. Dietary Antioxidant Deficiency Facilitates Cortical Spreading Depression Induced by Photoactivated Riboflavin. Nutr. Neurosci. 1998, 1, 205–212. [Google Scholar]
  111. Mandel, S.; Grünblatt, E.; Riederer, P.; Gerlach, M.; Levites, Y.; Youdim, M.B. Neuroprotective strategies in Parkinson’s disease: An update on progress. CNS Drugs 2003, 17, 729–762. [Google Scholar]
  112. Yu, Y.C.; Kuo, C.L.; Cheng, W.L.; Liu, C.S.; Hsieh, M. Decreased antioxidant enzyme activity and increased mitochondrial DNA damage in cellular models of Machado-Joseph disease. J. Neurosci. Res. 2009, 87, 1884–1891. [Google Scholar]
  113. Zeya, B.; Arjuman, A.; Chandra, N.C. Lectin-like Oxidized Low-Density Lipoprotein (LDL) Receptor (LOX-1): A Chameleon Receptor for Oxidized LDL. Biochemistry 2016, 55, 4437–4444. [Google Scholar]
  114. Kattoor, A.J.; Kanuri, S.H.; Mehta, J.L. Role of Ox-LDL and LOX-1 in Atherogenesis. Curr. Med. Chem. 2019, 26, 1693–1700. [Google Scholar]
  115. Yu, S.; Zhang, L.; Liu, C.; Yang, J.; Zhang, J.; Huang, L. PACS2 is required for ox-LDL-induced endothelial cell apoptosis by regulating mitochondria-associated ER membrane formation and mitochondrial Ca(2+) elevation. Exp. Cell Res. 2019, 379, 191–202. [Google Scholar]
  116. Nègre-Salvayre, A.; Augé, N.; Camaré, C.; Bacchetti, T.; Ferretti, G.; Salvayre, R. Dual signaling evoked by oxidized LDLs in vascular cells. Free Radic. Biol. Med. 2017, 106, 118–133. [Google Scholar]
  117. Lubrano, V.; Balzan, S. LOX-1 and ROS, inseparable factors in the process of endothelial damage. Free Radic Res. 2014, 48, 841–848. [Google Scholar]
  118. Maiolino, G.; Rossitto, G.; Caielli, P.; Bisogni, V.; Rossi, G.P.; Calò, L.A. The role of oxidized low-density lipoproteins in atherosclerosis: The myths and the facts. Mediat. Inflamm. 2013, 2013, 714653. [Google Scholar]
  119. Zhou, Y.D.; Cao, X.Q.; Liu, Z.H.; Cao, Y.J.; Liu, C.F.; Zhang, Y.L.; Xie, Y. Rapamycin Inhibits Oxidized Low Density Lipoprotein Uptake in Human Umbilical Vein Endothelial Cells via mTOR/NF-κB/LOX-1 Pathway. PLoS ONE 2016, 11, e0146777. [Google Scholar]
  120. Xu, S.; Ogura, S.; Chen, J.; Little, P.J.; Moss, J.; Liu, P. LOX-1 in atherosclerosis: Biological functions and pharmacological modifiers. Cell Mol. Life Sci. 2013, 70, 2859–2872. [Google Scholar]
  121. Kattoor, A.J.; Pothineni, N.V.K.; Palagiri, D.; Mehta, J.L. Oxidative Stress in Atherosclerosis. Curr. Atheroscler Rep. 2017, 19, 42. [Google Scholar]
  122. Civelek, M.; Manduchi, E.; Riley, R.J.; Stoeckert, C.J., Jr.; Davies, P.F. Chronic endoplasmic reticulum stress activates unfolded protein response in arterial endothelium in regions of susceptibility to atherosclerosis. Circ. Res. 2009, 105, 453–461. [Google Scholar]
  123. Hetz, C. The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat. Rev. Mol. Cell Biol. 2012, 13, 89–102. [Google Scholar]
  124. Rutkowski, D.T.; Kaufman, R.J. A trip to the ER: Coping with stress. Trends Cell Biol. 2004, 14, 20–28. [Google Scholar]
  125. Tabas, I. The role of endoplasmic reticulum stress in the progression of atherosclerosis. Circ. Res. 2010, 107, 839–850. [Google Scholar]
  126. Yang, S.; Wu, M.; Li, X.; Zhao, R.; Zhao, Y.; Liu, L.; Wang, S. Role of Endoplasmic Reticulum Stress in Atherosclerosis and Its Potential as a Therapeutic Target. Oxid Med. Cell Longev. 2020, 2020, 9270107. [Google Scholar]
  127. Seidlmayer, L.K.; Juettner, V.V.; Kettlewell, S.; Pavlov, E.V.; Blatter, L.A.; Dedkova, E.N. Distinct mPTP activation mechanisms in ischaemia-reperfusion: Contributions of Ca2+, ROS, pH, and inorganic polyphosphate. Cardiovasc. Res. 2015, 106, 237–248. [Google Scholar]
  128. Görlach, A.; Bertram, K.; Hudecova, S.; Krizanova, O. Calcium and ROS: A mutual interplay. Redox Biol. 2015, 6, 260–271. [Google Scholar]
  129. Duchen, M.R. Mitochondria and calcium: From cell signalling to cell death. J. Physiol. 2000, 529 Pt 1, 57–68. [Google Scholar]
  130. Martínez-Klimova, E.; Aparicio-Trejo, O.E.; Gómez-Sierra, T.; Jiménez-Uribe, A.P.; Bellido, B.; Pedraza-Chaverri, J. Mitochondrial dysfunction and endoplasmic reticulum stress in the promotion of fibrosis in obstructive nephropathy induced by unilateral ureteral obstruction. Biofactors 2020, 46, 716–733. [Google Scholar]
  131. Borutaite, V.; Toleikis, A.; Brown, G.C. In the eye of the storm: Mitochondrial damage during heart and brain ischaemia. FEBS J. 2013, 280, 4999–5014. [Google Scholar]
  132. Nakagawa, T.; Zhu, H.; Morishima, N.; Li, E.; Xu, J.; Yankner, B.A.; Yuan, J. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature 2000, 403, 98–103. [Google Scholar]
  133. Garrido, C.; Galluzzi, L.; Brunet, M.; Puig, P.E.; Didelot, C.; Kroemer, G. Mechanisms of cytochrome c release from mitochondria. Cell Death Differ. 2006, 13, 1423–1433. [Google Scholar]
  134. Bratton, S.B.; Salvesen, G.S. Regulation of the Apaf-1-caspase-9 apoptosome. J. Cell Sci. 2010, 123 Pt 19 Pt 19, 3209–3214. [Google Scholar]
  135. Jeong, S.Y.; Seol, D.W. The role of mitochondria in apoptosis. BMB Rep. 2008, 41, 11–22. [Google Scholar]
  136. de Oliveira, J.; Hort, M.A.; Moreira, E.L.; Glaser, V.; Ribeiro-do-Valle, R.M.; Prediger, R.D.; Farina, M.; Latini, A.; de Bem, A.F. Positive correlation between elevated plasma cholesterol levels and cognitive impairments in LDL receptor knockout mice: Relevance of cortico-cerebral mitochondrial dysfunction and oxidative stress. Neuroscience 2011, 197, 99–106. [Google Scholar]
  137. Han, Z.; Wang, Y.; Li, J. Effects of Atorvastatin Combined with Nano-Selenium on Blood Lipids and Oxidative Stress in Atherosclerotic Rats. J. Nanosci. Nanotechnol. 2021, 21, 1331–1337. [Google Scholar]
  138. Moreira, E.L.; de Oliveira, J.; Nunes, J.C.; Santos, D.B.; Nunes, F.C.; Vieira, D.S.; Ribeiro-do-Valle, R.M.; Pamplona, F.A.; de Bem, A.F.; Farina, M.; et al. Age-related cognitive decline in hypercholesterolemic LDL receptor knockout mice (LDLr−/−): Evidence of antioxidant imbalance and increased acetylcholinesterase activity in the prefrontal cortex. J. Alzheimers Dis. 2012, 32, 495–511. [Google Scholar]
  139. Casula, M.; Catapano, A.L.; Bernardi, L.R.; Visconti, M.; Aronica, A. Detection of familial hypercholesterolemia in patients from a general practice database. Atheroscler Suppl. 2017, 29, 25–30. [Google Scholar]
  140. El Shamieh, S.; Costanian, C.; Kassir, R.; Visvkis-Siest, S.; Bissar-Tadmouri, N. APOE genotypes in Lebanon: Distribution and association with hypercholesterolemia and Alzheimer’s disease. Per Med. 2019, 16, 15–23. [Google Scholar]
  141. Shi, Y.; Andhey, P.S.; Ising, C.; Wang, K.; Snipes, L.L.; Boyer, K.; Lawson, S.; Yamada, K.; Qin, W.; Manis, M.; et al. Overexpressing low-density lipoprotein receptor reduces tau-associated neurodegeneration in relation to apoE-linked mechanisms. Neuron 2021, 109, 2413–2426.e7. [Google Scholar]
  142. Mahley, R.W. Apolipoprotein E: Cholesterol transport protein with expanding role in cell biology. Science 1988, 240, 622–630. [Google Scholar]
  143. Rhea, E.M.; Banks, W.A. Interactions of Lipids, Lipoproteins, and Apolipoproteins with the Blood-Brain Barrier. Pharm. Res. 2021, 38, 1469–1475. [Google Scholar]
  144. Namba, Y.; Tomonaga, M.; Kawasaki, H.; Otomo, E.; Ikeda, K. Apolipoprotein E immunoreactivity in cerebral amyloid deposits and neurofibrillary tangles in Alzheimer’s disease and kuru plaque amyloid in Creutzfeldt-Jakob disease. Brain Res. 1991, 541, 163–166. [Google Scholar]
  145. Wisniewski, T.; Frangione, B. Apolipoprotein E: A pathological chaperone protein in patients with cerebral and systemic amyloid. Neurosci. Lett. 1992, 135, 235–238. [Google Scholar]
  146. Jordan, B.D.; Relkin, N.R.; Ravdin, L.D.; Jacobs, A.R.; Bennett, A.; Gandy, S. Apolipoprotein E epsilon4 associated with chronic traumatic brain injury in boxing. JAMA 1997, 278, 136–140. [Google Scholar]
  147. Mayeux, R.; Ottman, R.; Maestre, G.; Ngai, C.; Tang, M.X.; Ginsberg, H.; Chun, M.; Tycko, B.; Shelanski, M. Synergistic effects of traumatic head injury and apolipoprotein-epsilon 4 in patients with Alzheimer’s disease. Neurology 1995, 45 Pt 1, 555–557. [Google Scholar]
  148. Mannix, R.C.; Zhang, J.; Park, J.; Zhang, X.; Bilal, K.; Walker, K.; Tanzi, R.E.; Tesco, G.; Whalen, M.J. Age-dependent effect of apolipoprotein E4 on functional outcome after controlled cortical impact in mice. J. Cereb. Blood Flow Metab. 2011, 31, 351–361. [Google Scholar]
  149. Golde, T.E.; Schneider, L.S.; Koo, E.H. Anti-aβ therapeutics in Alzheimer’s disease: The need for a paradigm shift. Neuron 2011, 69, 203–213. [Google Scholar]
  150. Benilova, I.; Karran, E.; de Strooper, B. The toxic Aβ oligomer and Alzheimer’s disease: An emperor in need of clothes. Nat. Neurosci. 2012, 15, 349–357. [Google Scholar]
  151. Hardy, J.; Selkoe, D.J. The amyloid hypothesis of Alzheimer’s disease: Progress and problems on the road to therapeutics. Science 2002, 297, 353–356. [Google Scholar]
  152. Basak, J.M.; Verghese, P.B.; Yoon, H.; Kim, J.; Holtzman, D.M. Low-density lipoprotein receptor represents an apolipoprotein E-independent pathway of Aβ uptake and degradation by astrocytes. J. Biol. Chem. 2012, 287, 13959–13971. [Google Scholar]
  153. Castellano, J.M.; Deane, R.; Gottesdiener, A.J.; Verghese, P.B.; Stewart, F.R.; West, T.; Paoletti, A.C.; Kasper, T.R.; DeMattos, R.B.; Zlokovic, B.V.; et al. Low-density lipoprotein receptor overexpression enhances the rate of brain-to-blood Aβ clearance in a mouse model of β-amyloidosis. Proc. Natl. Acad. Sci. USA 2012, 109, 15502–15507. [Google Scholar]
  154. de Oliveira, J.; Moreira, E.L.; Santos, D.B.d.; Piermartiri, T.C.; Dutra, R.C.; Pinton, S.; Tasca, C.I.; Farina, M.; Prediger, R.D.; de Bem, A.F. Increased susceptibility to amyloid-β-induced neurotoxicity in mice lacking the low-density lipoprotein receptor. J. Alzheimers Dis. 2014, 41, 43–60. [Google Scholar]
  155. Zhao, Y.; Yang, Y.; Xing, R.; Cui, X.; Xiao, Y.; Xie, L.; You, P.; Wang, T.; Zeng, L.; Peng, W.; et al. Hyperlipidemia induces typical atherosclerosis development in Ldlr and Apoe deficient rats. Atherosclerosis 2018, 271, 26–35. [Google Scholar]
  156. Shi, Y.; Holtzman, D.M. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat. Rev. Immunol. 2018, 18, 759–772. [Google Scholar]
  157. Mulder, M.; Jansen, P.J.; Janssen, B.J.; van de Berg, W.D.; van der Boom, H.; Havekes, L.M.; de Kloet, R.E.; Ramaekers, F.C.; Blokland, A. Low-density lipoprotein receptor-knockout mice display impaired spatial memory associated with a decreased synaptic density in the hippocampus. Neurobiol. Dis. 2004, 16, 212–219. [Google Scholar]
Figure 1. Interaction of low-density-lipoprotein receptor (LDLr) and insulin receptor (IR). The process of the expression of LDLr and endocytosed downstream molecules to regulate cholesterol homeostasis and the association of co-localized IR.
Figure 1. Interaction of low-density-lipoprotein receptor (LDLr) and insulin receptor (IR). The process of the expression of LDLr and endocytosed downstream molecules to regulate cholesterol homeostasis and the association of co-localized IR.
Ijms 23 08384 g001
Figure 2. Blood–brain-barrier (BBB) breakdown due to inflammation. BBB breakdown process induced by NLRP3-mediated inflammation.
Figure 2. Blood–brain-barrier (BBB) breakdown due to inflammation. BBB breakdown process induced by NLRP3-mediated inflammation.
Ijms 23 08384 g002
Figure 3. Interaction between ER stress and mitochondria. The process by which ER stress and mitochondria interact due to oxLDL accumulation due to LDLr deficiency affects cognitive impairment.
Figure 3. Interaction between ER stress and mitochondria. The process by which ER stress and mitochondria interact due to oxLDL accumulation due to LDLr deficiency affects cognitive impairment.
Ijms 23 08384 g003
Figure 4. The process of the clearance of ApoE/Aβ complexes by LDLr.
Figure 4. The process of the clearance of ApoE/Aβ complexes by LDLr.
Ijms 23 08384 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hong, D.-Y.; Lee, D.-H.; Lee, J.-Y.; Lee, E.-C.; Park, S.-W.; Lee, M.-R.; Oh, J.-S. Relationship between Brain Metabolic Disorders and Cognitive Impairment: LDL Receptor Defect. Int. J. Mol. Sci. 2022, 23, 8384. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23158384

AMA Style

Hong D-Y, Lee D-H, Lee J-Y, Lee E-C, Park S-W, Lee M-R, Oh J-S. Relationship between Brain Metabolic Disorders and Cognitive Impairment: LDL Receptor Defect. International Journal of Molecular Sciences. 2022; 23(15):8384. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23158384

Chicago/Turabian Style

Hong, Dong-Yong, Dong-Hun Lee, Ji-Young Lee, Eun-Chae Lee, Sang-Won Park, Man-Ryul Lee, and Jae-Sang Oh. 2022. "Relationship between Brain Metabolic Disorders and Cognitive Impairment: LDL Receptor Defect" International Journal of Molecular Sciences 23, no. 15: 8384. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23158384

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop