Next Article in Journal
Alterations of Mitochondrial Structure in Methamphetamine Toxicity
Next Article in Special Issue
The Crosstalk between HepG2 and HMC-III Cells: In Vitro Modulation of Gene Expression with Conditioned Media
Previous Article in Journal
Characterization of the SWEET Gene Family in Longan (Dimocarpus longan) and the Role of DlSWEET1 in Cold Tolerance
Previous Article in Special Issue
Altered Retrograde Signaling Patterns in Breast Cancer Cells Cybrids with H and J Mitochondrial DNA Haplogroups
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Low-Density Lipoprotein Receptor-Related Protein 8 at the Crossroad between Cancer and Neurodegeneration

1
Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
2
Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, 90133 Palermo, Italy
3
Department of Medical Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100 Chieti, Italy
4
Department of Pharmacy, University of Chieti-Pescara, 66100 Chieti, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(16), 8921; https://doi.org/10.3390/ijms23168921
Submission received: 30 June 2022 / Revised: 7 August 2022 / Accepted: 9 August 2022 / Published: 10 August 2022
(This article belongs to the Special Issue Cancer and Neurodegenerative Diseases: What Is the Link?)

Abstract

:
The low-density-lipoprotein receptors represent a family of pleiotropic cell surface receptors involved in lipid homeostasis, cell migration, proliferation and differentiation. The family shares common structural features but also has significant differences mainly due to tissue-specific interactors and to peculiar proteolytic processing. Among the receptors in the family, recent studies place low-density lipoprotein receptor-related protein 8 (LRP8) at the center of both neurodegenerative and cancer-related pathways. From one side, its overexpression has been highlighted in many types of cancer including breast, gastric, prostate, lung and melanoma; from the other side, LRP8 has a potential role in neurodegeneration as apolipoprotein E (ApoE) and reelin receptor, which are, respectively, the major risk factor for developing Alzheimer’s disease (AD) and the main driver of neuronal migration, and as a γ-secretase substrate, the main enzyme responsible for amyloid formation in AD. The present review analyzes the contributions of LDL receptors, specifically of LRP8, in both cancer and neurodegeneration, pointing out that depending on various interactions and peculiar processing, the receptor can contribute to both proliferative and neurodegenerative processes.

1. Introduction

Cancer and neurodegeneration represent two age-related conditions that share common biochemical features, and it is not clear what triggers can lead to one or the other condition through apparently common routes and pathways [1,2,3]. It is well-known that the activation and deregulation of the cell cycle may lead to cell death in neurons and to uncontrolled proliferation in malignant cells; however, there is still a gap in the understanding of the precise molecular processes involved in both disorders; unraveling them represents an important opportunity to identify prognostic markers and to achieve therapeutic progress in both pathological conditions.
The low-density-lipoprotein receptors (LDLRs) represent a family of cell surface receptors originally linked to lipoprotein and lipid trafficking and, more recently, considered pivotal regulators of signal transduction pathways for cell proliferation, migration and differentiation [4]. Recent growing evidence has also highlighted their associations with neurodegenerative disorders and cancer due to the modulation of specific molecular pathways related to neuronal cell death and, in parallel, to cell proliferation, cell cycle activation and metastatic invasion [5,6,7,8,9].

2. LDLR Family

Listed in the order of their discovery, the main members of the LDLR family are: LDLR; LDLR-related protein (LRP, LRP1); Megalin (LRP-2, originally called gp330); very low-density lipoprotein (VLDL) receptor (VLDLR; in chicken termed LR8); LR11 (also named SorLA); apolipoprotein E (ApoE) receptor type 2 (LRP8, LRP-8) [4]; LRP-3, -4, -5, and -6, and LR32 (also termed LRP1B).
The structure of the LDL receptors, described in Figure 1, contains five common functional regions: a N-terminal ligand-binding domain with a variable number of LDLR type A (LA) repeats, an epidermal growth factor precursor homology (EGFP) domain composed of EGF-repeats (cysteine-rich class B repeats) and a YWTD (Tyr–Trp–Thr–Asp)/β-propeller domain, an O-linked sugar domain, a transmembrane domain and a cytoplasmic domain containing at least one NPxY (Asn-Pro-Xaa-Tyr) motif [10,11,12]. SorLA has two additional domains: the fibronectin type III (FNIII) domain and the vacuolar protein sorting 10 (VPS10) homology domain. In addition to the LDLR members, the NPXY motif is found in a subset of cell surface proteins well-known to be involved in cell proliferation and cancer including APP [13], growth factor receptors [14] and integrins [15].
The LDLR family participates in a wide range of physiological processes in different organs, tissues and cell types [4]. They are mainly known for binding and internalizing several extracellular ligands, including lipoproteins, exotoxins and lipid-carrier complexes, as well as for mediating signaling responses as a result of changes in the extracellular environment. The presence of the NPxY motif in the cytoplasmic domain represents an intracellular binding site for many adaptor proteins such as the mammalian Disabled-l (mDab-1), mDab-2, FE65, JNK-interacting protein JIP-1 and JIP-2, PSD-95, CAPON, and SEMCAP-1 [16,17,18]; many of these contain in their structure at least one of the three frequently found interaction domains such as phosphotyrosine binding (PTB), PDZ or SH2-SH3 domains or ankyrin repeats.
A functional peculiarity of the LDL receptors, especially LRP8, is their double functional role in neurodegeneration and cancer, the main focus of this review, as summarized in Table 1.

2.1. LDLR

The low-density lipoprotein receptor (LDLR) is the prototype of a classical endocytosis receptor that mediates the uptake of ligands and is broadly expressed on multiple cell types in various tissues. In the CNS, the LDLR plays an important role in regulating the homeostasis of cholesterol in the blood and also within tissues and cells [19]. The main well-known LDLR ligand is represented by apolipoprotein E (ApoE), a 34 kDa soluble protein that is a main component of lipoproteins in plasma and a secreted glycoprotein of 34 kDa that acts as a cholesterol carrier and signaling molecule. Through its interaction with LDLR, ApoE plays a critical role in peripheral cholesterol metabolism [20] and also in cholesterol transport into the central nervous system (CNS) [21,22]. LDLR is the only member of this receptor family to demonstrate an apoE isoform-specific binding affinity (E4 > E3 >> E2). Evidence suggests that a functional interaction between ApoE and LDLR influences the risk of cardiovascular disease (CVD) and AD [23]. The apoE–LDLR interaction may also play a more direct role through the modulation of amyloid clearance and/or deposition. Several intriguing studies have shed some light on the potential pathophysiological pathways by which LDLR may contribute to AD. In cell culture, overexpression of the amyloid precursor protein (APP) led to the increased expression of LDLR and altered receptor localization [24]. Varying results in amyloid deposition have been shown in AD mouse models lacking the LDLR, with one study showing that amyloid deposition was enhanced on a LDLR null background [25], while in another study, LDLR deficiency had no effect on brain amyloid-β peptides (Aβ) levels [26].
Kim et al., showed that LDLR over-expression reduced brain ApoE levels and amyloid β aggregation [27], while further work from the same laboratory showed that LDLR over-expression in the brain increased the rate of brain-to-blood clearance of both exogenously administered and endogenous Aβ [28]. These studies, in which LDLR over-expression increases Aβ clearance, point to the upregulation of LDLR in the brain as a potential therapeutic strategy for AD.
Considering the vital role of the LDL/LDLR routine in regulating blood and intracellular cholesterol homeostasis, several studies have focused on the function of LDLR in cancer progression. Chen et al., have demonstrated that the reduced expression of LDLR in hepatocellular carcinoma cells impairs LDL uptake but promotes proliferation and metastasis in vitro and in vivo by activating MEK/ERK [29].

2.2. LRP1

LRP1 represents a key receptor in maintaining lipid metabolism. The expression of LRP1 is ubiquitous, and its upregulation has been reported in numerous human diseases. In addition to its function as a scavenger receptor for various ligands, LRP1 transduces multiple intracellular signal pathways including mitogen-activated protein kinase (MAPK), Akt, Rho and the integrin signaling pathway. LRP1 signaling plays an important role in the regulation of diverse cellular processes, such as cell proliferation, survival, motility, differentiation and transdifferentiation, and thus, it participates in the pathogenesis of organ dysfunction and injury [30]. LRP1 is widely expressed throughout the CNS, where it modulates neuronal survival, neurite outgrowth, regeneration and calcium signaling through different ligands and cell-specific mechanisms. Indeed, LRP1 recognizes and is involved in the endocytosis of more than 40 different ligands including ApoE, APP and Aβ [31,32,33]. Depending on its binding to different ApoE isoforms, LRP1 is involved in calcium-related cellular processes such as increasd resting calcium, calcium response to N-methyl-D-aspartate (NMDA) and neurotoxicity [34]. Moreover, LRP1 binding to lipoprotein particles containing ApoE 3/4 protects against apoptosis upon the activation of protein kinase Cδ (PKCδ) and the inactivation of glycogen synthase kinase-3β (GSK3β), with a greater effect due to lipoproteins with ApoE4 than those with ApoE 3 [35]. The majority of research examining LRP1 function in the brain has focused on APP trafficking [36,37], Aβ clearance from the brain parenchyma [38] and blood–brain barrier permeability [39]. Studies on knockout mouse models have shown that LRP1 deficiency is lethal early in development [40], whereas mice lacking LRP1 exhibit severe motor and behavioral abnormalities, hyperactivity, age-dependent dendritic spine degeneration, synapse loss, neuroinflammation, memory loss and eventual neurodegeneration [41,42]. In addition to its role in neuronal-related physiological processes, an additional role in cancer development has been associated with LRP1. In particular in human glioblastoma U87 cells, LRP1 promotes cellular migration and invasion by inducing the expression of the matrix metalloproteinase-2 (MMP-2) and MMP-9 [43]. Moreover, in U87 cells, Song et al., have proposed that phosphorylated ERK is a potential mediator of LRP1-regulated MMP expression. An additional piece to this puzzle has been provided by Fayard et al., suggesting that the serine protease inhibitor serpin PN-1 is a ligand of LRP1 that activates ERK and stimulates, in turn, MMP-9 expression in breast tumor cell lines [44]. Supporting this hypothesis, an analysis of 126 breast cancer patients revealed that those patients whose breast tumors had elevated PN-1 levels had significantly higher probabilities of developing lung metastasis, but not metastasis to other sites, on relapse. Finally, it has been found that in Her-2/neu and triple-negative breast cancer (TNBC), there is an increased expression of LRP1 that is linked to neoplastic aggressiveness due to high histological grade, elevated mitotic index, Ki67 > 20% and tumor recurrence, while in endometrial carcinoma (EC), the increased expression of LRP1 is associated with p53 alterations and p16 protein overexpression [45].

2.3. LRP2/Megalin

LRP2 is a multiligand receptor mostly localized in the epithelial cells of the renal tubules [46] and the choroid plexus [47], as well as in the lateral ventricles [48] and in different CNS cells ranging from oligodendrocytes, retinal ganglion cells, cerebellar granule neurons and astrocytes to hippocampal neurons [49]. In the CNS, LRP2 modulates neuronal survival and regeneration [50] as well as learning- and memory-related physiological mechanisms such as neurite outgrowth and synaptic plasticity [49]. LRP2 neurotrophic activity is performed through its binding with Transthyretin [51], while its neurite outgrowth function seems associated with metallothionines [52] and the APP/Fe65 pathway [53]. In the blood–brain barrier, LRP2 plays a central role in the clearance/entrance of many proteins from the brain/cerebrospinal fluid, including Aβ, insulin, insulin-like growth factor (IGF)-I and ApoE. Together with LRP8, LRP2 is also known to transport selenium, which is transported in the form of the selenocysteine-enriched selenoprotein P1 (Sepp1) to target tissues, where it constitutes the active center of glutathione peroxidase, thioredoxin reductase and deiodinases. In the kidney, proximal tubule epithelial cells highly express LRP2, indicating its physiological role in the reabsorption of Sepp1. This function is confirmed by studies on LRP2-mutant mice displaying urinary selenium loss, which correlates with Sepp1 excretion in their urine and reduced selenium and glutathione peroxidase activity in the brain [54].
LRP2 genetic alterations have also been associated with pathological conditions. In particular, some LRP2 mutations lead to the protein loss of function, underlying an autosomal recessive disorder, Donnai–Barrow syndrome, characterized by several CNS functional defects [55]. Other genetic studies have associated a polymorphism located in the LRP2 promoter with AD risk, regardless of the ApoE genotype [56], as well as some LRP2 germ-line polymorphisms with increased risk of prostate cancer recurrence [57]. Moreover, somatic LRP2 mutations have been identified in gastric cancer [58]. In support of LRP2’s role in cancer, Andersen et al., have reported that LRP2 is more frequently expressed among malignant melanoma samples compared with benign counterparts and that the proliferation and survival rates of cultured LRP2-expressing melanoma cells decrease upon the siRNA-mediated knockdown of LRP2 [7].

2.4. VLDLR

VLDLR is highly expressed in adipose tissue, heart and skeletal muscle and is mainly known for the following roles: uptake of VLDL-derived lipids, enhancement of lipoprotein lipase activity and mediation of the Reelin-related pathway, together with LRP8 [59]. Reelin binding to VLDLR/LRP8 receptors activates intracellular Src family kinases (SFKs), which in turn phosphorylate the adaptor protein Disabled-1 (Dab1) in specific tyrosine residues. Although the activation of the Reelin pathway through VLDLR and LRP8 is well-known to mediate the development of the central nervous system, studies have highlighted differences between the two receptors in terms of splicing, localization, interactors and trafficking, leading to unique functions separate from the overlapping ones. In particular, the Pafah1b complex mediates the downstream effects of VLDLR on neuronal migration, but it is not necessary for the function of LRP8 [60].
In addition to its roles in neuronal physiological processes, studies have highlighted the role of VLDLR in cancer. Indeed, Lei He et al., have shown that VLDLR promotes cell proliferation and migration in adenocarcinoma SGC7901 cells via beta-catenin/TCF signaling [8], and Lei He et al., have correlated VLDLR II expression with lymph node and distant metastasis in gastric and breast cancer patients [61].

2.5. LRP4

Although it is mainly expressed in bone, where it regulates bone formation by inhibiting sclerostin in Wnt1/β-catenin signaling [62], LRP4 is also found in astrocytes of the prefrontal cortex and hippocampus, regions particularly vulnerable to AD [63]. Studies of the genetic deletion of the Lrp4 gene in 5xFAD male mice, an AD mouse model, have shown increased levels of Aβ plaques and decreased neurotransmission and cognition, supporting a link between LRP4 loss and AD [64]. In the brain, LRP4 plays roles in synaptic transmission, long-term potentiation (LTP), cognitive function [65] and adult hippocampal neurogenesis [66]. Through its ligand agrin, LRP4 regulates the formation and maintenance of the neuromuscular junction [67,68]. In particular, the agrin–Lrp4 interaction increases its binding with MuSK, which is activated and thus activates the kinase that via Dok7 and rapsyn mediates AChR clustering [69]. In addition to AD, LRP4 has been associated with cancer. In papillary thyroid cancer, LRP4 has been reported to be significantly overexpressed in both tissues and related cell lines (TPC1, BCPAP and KTC-1) in comparison with controls, likely playing a role in proliferation, migration and invasion by inducing phosphoinositide 3-kinases (PI3K)/AKT mediated epithelial–mesenchimal transition (EMT) [70]. LRP4 overexpression has also been observed in gastric cancer tissues, where it correlates with malignant proliferating clinical features, while in gastric cancer cells (MKN45, MGC803, BGC823 and AGS), LRP4 activates the PI3K/AKT under regulation of miR-140-5p [71].

2.6. LR11/SorLA

SorLA is highly enriched in the brain. As LRP1, SorLA interacts with APP, influencing its trafficking and regulating Aβ levels [72,73]. Indeed, SorLA represents one of the major cellular players contributing to abnormal APP processing and enhanced Aβ formation. In particular, SorLA acts as a retention factor for APP in trans-Golgi compartments/trans-Golgi network, with GGA and PACS-1 adaptor proteins involved in protein transport to and from the trans-Golgi network, preventing the release of the precursor into regular processing pathways [74]. Interestingly, a peculiar alternative splicing of SORL1, induced by a non-coding RNA, is associated with the impaired processing of APP and Aβ formation [75]. A consistent loss of SorLA is observed in AD brains, particularly in vulnerable regions [76], and variants in the SORL1 gene encoding SorLA have been associated with AD risk, adding considerable support to the hypothesis that SorLA is genetically linked to AD pathogenesis [77]. However, SorLA represents another LDLR whose overexpression is linked to cancer, and in particular, it has been observed in HER2-driven cancer cell lines where the receptor regulates endosomal trafficking and oncogenic fitness of HER2 [78]. SorLA and its released soluble form are also strongly elevated in acute leukemia. Remarkably, the increased levels of soluble SorLA level were reduced in patients who achieved complete remission [79,80].

2.7. LRP5/6

LRP5 and LRP6 are two LDL receptors with amino acid sequences with 71% identity [81]. Their extracellular domains are responsible for binding Wnt ligands and their inhibitors, such as Dickkopf-related protein 1 (DKK1) and sclerostin [82]; indeed, LRP5 and LRP6 are the key players that the Wnt pathway together with the Frizzled receptors [83,84,85]. In particular, Wnt ligands signal through at least three different pathways: the canonical or Wnt/β-catenin pathway and the two noncanonical Wnt/JNK and Wnt/Ca2+ pathways. In the canonical pathway, the Wnt-Fzd-LRP5/6 leads to a downregulation of glycogen synthase kinase-3 (GSK-3) activity that in turn increases β-catenin levels in the cytosol, thereby facilitating its translocation to the nucleus. At the nuclear level, β-catenin forms complexes with members of the Tcf/Lef class of DNA-binding proteins modulating transcriptional activity of target promoters [86,87]. The Wnt-Fzd-LRP5/6 interaction leads to the inhibition of the axin degradasome destruction complex that is recruited to the plasma membrane, helping the interaction between LRP5/6 and axin. When LRP5/6 is phosphorylated at specific amino acidic residues (Ser1490, Thr1530, Thr1572, Ser1590, Ser1607), it acts as a direct competitive inhibitor of GSK3. The Wnt signaling, most probably through LRP5 and LRP6, can also directly activate the mammalian target of rapamycin (mTOR) pathway by decreasing GSK-3-mediated activation of the TSC2/TSC1 complex. The activation of mTOR signaling promotes cell growth in addition to representing a vital regulator of autophagy [88].
LRP5 and LRP6 receptors are expressed differently in various tissues and organs. In humans, LRP5 expression level is the highest in liver, while substantial level of expression is also observed in pancreas, prostate, placenta and small intestine. The expression level of this receptor in brain and peripheral leukocytes is very low [89]. Human LRP6 expression is highest in the ovary, with significant levels in the heart, brain, placenta, lung, kidney, pancreas, spleen and testis. The expression of LRP6 is very low in peripheral blood leukocytes, thymus and small intestine [81].
LRP5 and LRP6 have been shown to play important roles in a broad panel of cancers. This is supported by the fact that the main pathway activated by both receptors is the Wnt/β-catenin pathway, which represents a pleiotropic signal involved in cell growth, cell proliferation and polarity, cell differentiation and tissue development [90,91] with a potential link to several diseases including cancer [92,93,94].
LRP5 has been reported to be involved in mediating Wnt/β-catenin signaling in skeletal metastasis prostate cancer (PC) due to the increased levels of Wnt-1 and β-catenin proteins in both PC cell lines and primary specimens [95]. The presence of LRP5 has also been linked significantly with tumor metastasis such as the chondroblastic subtype of osteosarcoma [96].
LRP6 overexpression, observed in many types of cancer and malignant tissues, leads to an abnormal Wnt pathway that is linked to tumorigenesis [97]. In humans, some LRP6 single-nucleotide polymorphisms (SNPs) and mutations in the LRP6 gene have been associated with increased or decreased risk of cancer development [98]. For example, an LRP6 variant (rs6488507) in non-small-cell lung cancer (NSCLC) patients is linked to an increase in the risk of NSCLC in tobacco smokers [99]. Moreover, the overexpression of LRP6 has been documented in breast cancer [97], hepatocellular carcinoma [100], colorectal cancer [101] and pancreatic ductal adenocarcinoma (PDAC) KRAS-dependent pancreatic cancer [102]. Consistent with these observations, a reduction of LRP6 expression and/or activity inhibits cancer cell proliferation and delays tumor growth in vivo [98,103]. Although mainly known as a co-receptor of the Wnt signaling pathway and therefore related to proliferative cellular pathways, some studies have described a connection of LRP6 to neurodegeneration. The LRP6-mediated Wnt signaling pathway is, in fact, compromised in AD brains, and its deficiency in an AD mouse model exacerbates amyloid pathology, synapse loss and Aβ toxicity, synergistically accelerating AD progression [104].

2.8. LRP1B

LRP1B is closely related to LRP1 due to their overlapping structural features and thus have shared ligands such as complexes of urokinase plasminogen activator, plasminogen activator inhibitor type-1 and receptor-associated protein (RAP) [105]. Both uPA and PAI-1 are key components of the uPA system, one of the major extracellular matrix-degrading proteinase systems playing a central role in cancer invasion and metastasis as well as other physiological and pathological processes involved in tissue remodeling [106]. LRP1B ligands link the receptor to altered cellular invasion/metastasis and support the correlations observed between LRP1B genetic and epigenetic alterations and many types of cancer [107,108,109], including urothelial malignancies [110], esophageal squamous cell carcinomas [111], gliomas, cervical adenocarcinomas, B-cell lymphomas [112], leukemias [113] and primary pulmonary adenocarcinomas [114]. In particular, its inactivation in 40% of NSCLC cell lines caused this receptor to initially be named LRP-deleted in tumors (LRP-DIT) [115]. Despite its structural link to LRP1, however, LRP1B differs from LRP in terms of tissue expression, localizing mainly in the brain (cortex, hippocampus and cerebellum in neurons, activated astrocytes and microglia), thyroid and salivary gland [105,116,117]. Its wide expression in the brain may suggest a yet-unclear CNS-related biological function, in part supported by its link to Aβ generation. In fact, Cam et al., have shown that LRP1B decreases APP internalization, thus reducing Aβ generation in a framework of anti-amyloid activity [118].
Table 1. CNS/neurodegeneration and the cancer-related roles of the LDL receptors. The table summarizes roles and relative molecular pathways of all LDLR (except LRP8, which is discussed later in detail) in CNS/neurodegeneneration and cancer.
Table 1. CNS/neurodegeneration and the cancer-related roles of the LDL receptors. The table summarizes roles and relative molecular pathways of all LDLR (except LRP8, which is discussed later in detail) in CNS/neurodegeneneration and cancer.
LDLR Family Members CNS Roles/
Neurodegeneration
Molecular PathwayRefs.Cancer Related RolesMolecular
Pathway
Refs.
LDLRModulation of amyloid clearance and/or depositionApoE interaction[25,27,28]Downregulation
of cell
proliferation and metastasis
Downregulation of
LDLR through
MEK/ERK stimulation
[29]
LRP1Neuronal
survival
ApoE-dependent activation of PKCδ and inactivation of GSK3β[35]
APP trafficking regulation and processing and
Aβ clearance
LRP1 antagonist RAP increases cell surface levels of APP and significantly reduce Aβ synthesis.
In the absence of LRP1, Aβ production, APP secretion,
APP internalization,
turnover of full-length APP
and stability of APP
C-terminal fragments are affected.
At the site of the BBB, surface LRP1-mediated extrusion of cerebral Aβ into the luminal side
Soluble LRP1 in the periphery sequesters free Aβ in circulation.
[36,37,38,39]Cellular migration
and invasion
Expression of MMP-2 and MMP-9 through ERK in human glioblastoma
Serpin PN-1-dependend MMP-9 expression through ERK activation in breast cancer
[43,44]
Calcium-related cellular processesApoE4, but not ApoE3, significantly increased the resting calcium,
the calcium response to NMDA-R and the neurotoxicity.
[34]Cell proliferation, tumor invasion and angiogenesisLRP1 expression has been linked to neoplastic aggressiveness due to high histological grade and elevated mitotic index.
Regeneration of the uPAR receptor system
[45]
Neurite outgrowth, synaptic plasticity, learning and memory
modulation
Upon TTR binding to LRP2,
Src, NMDA-Rs, ERK1/2,
CREB and Akt activation
and/or a pathway
involving RIP and the
formation of LRP2-ICD
MT-IIA binding to LRP2 stimulates neurite outgrowth via signal transduction pathways activated by the NPxY motifs of LRP2.
[49,52]Cell survival and proliferationLRP2 is frequently expressed in malignant melanoma.
Modulation of phosphorylated Akt and ERK levels
[7]
VLDLRRegulation of the migration and layering of the neurons in the cortex and the cerebellumReelin-induced Dab1
binding to VLDLR
activates SFK and Abl families, together with LRP8.
Pafah1b complex mediates downstream effects of VLDLR on neuronal migration.
[59,60,119]Cell proliferation, migration and metastasisVLDLR II is overexpressed in lymph node and distant metastasis in gastric and breast cancer patients, promoting cell proliferation and migration.
ATRA attenuates proliferation and
migration through significant decreases in VLDLR II, while PMA has the opposite effect on VLDLR II, which activates β-catenin/TCF signaling and modulation of MMP-2 and MMP-9.
[8,61]
LRP4Synaptic homeostasisLRP4 mutant astrocytes suppressed glutamatergic transmission by enhancing the release of ATP.[63]
Synaptic transmission, LTP and cognitive functionLRP4 KO shows deficits in cognitive tasks with
aberrant synapse form
and function and loss of LTP.
[65]EMT promotionLRP4 is overexpressed in papillary thyroid and gastric cancers, where it promotes EMT through PI3K/AKT pathway
and modulation of N-cadherin, ZEB1 and EZH2.
[70,71]
Adult hippocampal neurogenesisLRP4 mutation blocks
NPSC proliferation.
Agrin-LRP4-Ror2 signaling
is involved in NSPC proliferation.
[66]Cell proliferation, migration and invasionLRP8 downregulation affects colony formation and migratory and invasive capacities through PI3K/AKT pathway.
miR-140-5p negatively regulates LRP4.
[70,71]
Formation and maintenance of the neuromuscular junctionAgrin–LRP4 interaction via MuSK, Dok7 and rapsyn mediates AChR clustering.[67,68]
LR11/
SorLA
Regulation of APP processing
and Aβ levels
Interaction with APP, enhancement of
APP in endosomal compartments and Golgi,
modulation of APP processing and reduction of Aβ levels
[72,73]Cell proliferationRegulation of endosomal trafficking and oncogenic fitness of HER2, promoting PI3K-dependent HER2 signaling[78]
LRP6Synaptic function
and integrity
Activation of
Wnt signaling
[104]Cell proliferation, survival and differentiation,
tumor growth
Co-receptor for
WNT and Wnt activator
[98,103]
LRP1BRegulation of APP endocytic rate and Aβ levels reductionInteraction with APP[118]Suppression of cell growth, invasion, migration, colony and tumor formationReduction of matrix
metalloproteinase 2 level
and negative regulation of uPAR
DNA methylation
[106,107,108]
Aβ, amyloid beta; BBB, blood-brain barrier; MMP-2, matrix metalloproteinase-2; MMP-9, matrix metalloproteinase-9; NMDA-R, N-methyl-D-aspartate receptor; uPAR, urokinase-type plasminogen activator receptor; MAP, mitogen-activated protein; CREB, cAMP response element-binding protein; MT, metallothionein; RIP, regulated intramembrane proteolysis; ICD, intracellular domain; SFK, Src kinase family; ATRA, All-trans retinoic acid; PMA, phorbol-12-myristate-13-acetate; LTP, long term potentiation; EMT, epithelial-mesenchymal transition; NPSCs, neural progenitor stem cells; AChR, acetylcholine receptor; PI3K, phosphoinositide 3-kinases; MuSK, muscle-specific Tyr kinase.

3. Apolipoprotein E Receptor 2 (LRP8)

LRP8 (gene name LRP8) is a modular type I transmembrane receptor of the LDLR family whose structure is made of one N-terminal extracellular ligand-binding domain made of seven conserved LA repeats, an EGF domain made of three EGF repeats and one β-propeller and one NPxY motif in the intracellular domain, as described in Figure 1 [120]. LRP8 is involved in many intracellular signaling pathways and cellular responses involving different ligands and co-receptors that will be discussed in detail in Section 3.1 and Section 3.2 and in the respective Figure 2 and Figure 3. The intracellular signaling pathways mediated by LRP8 mostly involve the carboxy-terminal NPXY motif [121,122]. This motif adopts in fact a tight hairpin conformation required for binding to phospho-tyrosine binding (PTB) domains or to SH2-SH3 domain-containing proteins. Ligand binding to LRP8, together with different LRP8 spliced variants, also modulates its proteolytic processing that releases C-terminal fragments (CTF) and a transcriptionally active intracellular domain (ICD) [123,124]. A detailed molecular mechanism of LRP8 proteolitic processing is described in Section 3.2.
As mentioned above, LRP8 has several splice variants, expressed in a tissue-specific manner [125,126,127], whose alteration has also been correlated to sporadic Alzheimer’s disease [128], that modify LRP8 functions such as ligand-binding activity [129], receptor glycosylation and processing [130] and downstream signaling [131]. A well-studied LRP8-spliced variant is the LR7/8B, which harbors high ligand-binding repeats and is known to be a receptor for α-macroglobulin together with LRP1 [132]. Another spliced variant, reported in all placental mammals so far studied and not in marsupials, birds or reptiles, includes exon 19 that encodes for a 59 amino acid proline-rich insert in the cytoplasmic domain of LRP8, which is unique in the LDLR family [133,134]. The human LRP8 unique aminoacidic sequence contains two potential SH3 binding motifs, PXXP (two prolines spaced by two other amino acids), suggesting a role in signal transduction pathways involving reelin, PSD-95, NMDA-R and JIPs and the corresponding functional roles in memory and spatial learning [16,18]. Indeed, its deletion in the mouse LRP8 gene (exon 19) explains defects in long-term memory storage and spatial learning, most probably due to the interruption of the reelin-mediated activation of NMDA receptors [134].
Additionally, there are variants missing epidermal growth factor repeat B or the O-glycosylation domain [126]. It has been reported that differential LRP8 splicing, together with its glycosylation at the O-linked sugar, regulates the release of the ICD. In this context, Wasser et al., have reported an alternative LRP8 splice variant lacking the O-linked glycosylation region that is cleaved more efficiently than the full-length counterparts [130]. More recently, it has been shown that LRP8 isoforms with differing numbers of ligand-binding repeats to generate different amounts of CTFs compared with full-length LRP8 [135].
LRP8 is enriched in the brain, in particular in the neocortex, cerebellum, hippocampus and olfactory bulb [136], and to a minor extent also in the peripheral nervous system such as in the sciatic nerve and Schwann cells [137]. Outside the nervous system, LRP8 can be found in placenta, testis, ovary and platelets and also in immune cells and vascular endothelial and smooth muscle cells [120,138]. Unlike LRP1, LRP8 inactivation has no impact on plasma triglyceride or cholesterol levels. Hence, the association between polymorphisms in the LRP8 gene and CVD risk is likely independent of lipid metabolism but related to LRP8-modulating cellular functions in the vessel wall [139].

3.1. LRP8 and Cancer

Although, LRP8 is mainly recognized for its role in CNS-related pathways, scientific evidence also suggests an involvement of the receptor in carcinogenesis. Generally, cancer cells require higher uptake of cholesterol than normal cells in which the receptor-mediated endocytosis of serum LDL enhances the cholesterol content through LDLR. Increases in LDLR and elevated plasma low-density lipoprotein cholesterol (LDL-C) have been reported as features of leukemia, glioblastoma and lung and pancreatic tumors [140]. This is explained by the fact that tumor cells rely on cholesterol for membrane and lipid raft biosynthesis, signaling molecules and other factors in order to meet the fast growth.
In this context, many studies summarized in Table 2 have highlighted the role of LRP8 in the tumorigenesis and progression of several cancers such as osteosarcoma, breast cancer, gastric cancer, hepatocellular carcinoma, lung cancer, prostate cancer and pancreatic cancer [6].
In fact, LRP8 is overexpressed in different cancers including osteosarcoma [141] and NSCLC [148], and its overexpression has been significantly correlated with poor clinicopathological features and prognosis. Abnormal LRP8 expression has also been associated with breast cancer progression, where it facilitates cell growth and confers a poor prognosis in patients. In particular, Maire et al., demonstrated that LRP8 is more strongly expressed in breast cancer without hormone receptor expression (TNBC and HER2 positive) than in luminal tumors (Luminal A and Luminal B) and that LRP8 depletion promotes apoptosis and impaired cell proliferation and colony formation. These findings have been further confirmed in an in vivo xenograft model where LRP8 depletion slowed tumor growth [142]. Other studies indirectly support the idea of a role of LRP8 in tumorigenesis: Dun et al., have shown that mycophenolic acid severely downregulates the expression of cell surface LRP8, inhibiting cell migration and the invasion of gastric cancer cells [143]; Cai et al., have separately found that LRP8 is responsible for hepatocellular carcinoma cell resistance to sorafenib [145].
In a scenario in which LRP8 seems to have an oncogenic role in many types of cancer, it remains still elusive the biochemical pathways through which LRP8 exerts such activity (Figure 2).
Figure 2. LRP8 in four main cancer-related molecular pathways. (1) LRP8 binding to ApoE2 induces ERK1/2 phosphorylation and cell cycle activation [150]. (2) LRP8 promotes Wnt-induced β-catenin accumulation, inducing Axin2 transcription [151]. (3) PCSK9 modulates LRP8 endocytosis and signaling by targeting the receptor for lysosomal degradation [152]. (4) LRP8 activates p-STAT3 and its nuclear signaling [141,153].
Figure 2. LRP8 in four main cancer-related molecular pathways. (1) LRP8 binding to ApoE2 induces ERK1/2 phosphorylation and cell cycle activation [150]. (2) LRP8 promotes Wnt-induced β-catenin accumulation, inducing Axin2 transcription [151]. (3) PCSK9 modulates LRP8 endocytosis and signaling by targeting the receptor for lysosomal degradation [152]. (4) LRP8 activates p-STAT3 and its nuclear signaling [141,153].
Ijms 23 08921 g002
One hypothetical mechanism is linked to ApoE isoform 2, which is known to have a critical role in cell proliferation and has been identified as a cancer-related molecule. Through its binding to LRP8, ApoE2 induces ERK1/2 phosphorylation, activating c-Myc that in turn inhibits p21Waf1 and thus removing the brake on the expression of cycle-related proteins such as cyclin D1, cdc2 and cyclin B1 [150].
In osteosarcoma, the overexpression of LRP8 compared with normal tissues, increases phospho-STAT3 (p-STAT3) levels and facilitates its translocation into the nucleus, resulting in an enhancement of mRNA, protein and promoter activity of the programmed death-ligand 1 (PD-L1), which is a marker of metastasis and mortality risk in osteosarcoma [141].
Among the intracellular signaling pathways positively regulated by LRP8, the Wnt/β-catenin is definitely one that links LRP8 to its tumorigenic role [151]. As already reported above, the Wnt/β-catenin pathway is a pleiotropic signal linked to several diseases including cancer. In particular, LRP8 seems to be a positive regulator of the Wnt pathway, increasing Wnt-induced transcriptional responses and promoting Wnt-induced β-catenin accumulation. The LRP8 regulation of the Wnt/β-Catenin pathway has been observed both in triple-negative breast cancer, where LRP8 depletion inhibits breast cancer stem cells, cell proliferation and invasion and epithelial–mesenchymal transition [9], and in NSLC, in which an increase in LRP8 expression facilitates tumor proliferation [148]. Furthermore, in KS483 osteoprogenitor cells, knockdown of LRP8 results in a decrease in the Wnt/β-Catenin pathway, with reduced β-catenin levels and the suppression of Axin2 transcription; as a secondary effect, the depletion of LRP8 decreases osteoblast differentiation and mineralization, whereas LRP8 ectopic expression has the opposite effect [151].
Together with VLDLR and LRP1, LRP8 is also involved in tumor cell growth through its binding to the glycoprotein Proprotein convertase subtilisin/kexin type 9 (PCSK9). PCSK9 is known to modulate cholesterol metabolism through its binding to LDLR family members, including LRP8, and promoting their degradation in intracellular acidic compartments. The binding between PCSK9 and the LDLR members occurs between the PCSK9 catalytic subunit and the LDLR EGF-A domain [152]. In this context, in addition to being a key player in the LDL metabolism in the liver and in the brain, PCSK9 is overexpressed in various human cancer cell lines [154].
Komaravolu et al., reported a cell-cycle modulation, not strictly related to cancer, that was mediated by LRP8 through its intracellular interaction with the catalytic subunit of the heterotrimeric enzyme PP2A, a protein complex that is vitally important for the maintenance of normal cell division [155,156]. In this regard, LRP8 participates in mitosis via its interaction with PP2A-C, promoting the formation of active CDC20 to complete cytokinesis. Moreover, since the LRP8-interacting adaptor protein Dab2 is also a substrate for PP2A dephosphorylation, LRP8 may work in concert with Dab2 to modulate cell cycle progression and cytokinesis [156].
LRP8 is also a key mediator in metastatic melanoma, as an ApoE receptor, in parallel with VLDLR and LRP1. According to Pencheva et al., selected miRNAs (miR-1908, miR-199a-5p and miR-199a-3p) limit ApoE secretion, thus suppressing its anti-metastatic action through the engagement of endothelial LRP8 receptor; in this condition, LRP8 no longer suppresses cell invasion and endothelial recruitment, eventually promoting metastasis [146].
Consistent with the above observations, LRP8 was also identified as a target of miR-30b-5p in lung cancer progression and cisplatin resistance. In particular, LRP8 level was markedly increased in lung cancer tissues compared with the control group, apparently acting as an oncogene. In addition, miR-30b-5p inhibits lung cancer cell viability, migration and invasion and enhances cell sensitivity to cysplatin via targeting LRP8, suggesting that miR-30b-5p inhibits lung cancer progression by targeting LRP8 [147]. Lu et al., published a similar work highlighting the role of miR-142 as a gastric cancer suppressor by targeting LRP8 [144]. A similar LRP8 regulation by miR-455-5p has been reported by Arai at al. in prostate cancer [149].

3.2. LRP8 in Neurodegeneration

LRP8 has long been studied for its role in cholesterol transport and metabolism; however, the identification of ApoE4, the major genetic risk factor for developing late-onset Alzheimer’s disease, as one of its ligands has brought attention to its role in CNS-related pathways and neurodegeneration (Figure 3).
Figure 3. LRP8 in CNS related molecular pathways and neurodegeneration. Ligands binding to LRP8 modulate intracellular pathways, LRP8 cleavage, LRP8-APP interaction and Aβ production. (1) ApoE. Aβ production is increased by ApoE binding to LRP8 [157]. (2) F-Spondin. F-Spondin increases LRP8 cleavage and decreases Aβ production [158]. (3) LRP8 levels also modulate Aβ production in a positive manner by increasing the APP association with lipid rafts and γ-secretase activity [159]. (4) Reelin. Reelin binding to LRP8 induces different signaling pathways: (a) the tyrosin phosphorylation of Dab1 by Src-family kinases [160] and its interaction with the LRP8 NPxY motif. Phosphorylated Dab1 subsequently interacts with PI3K, whose activation leads to the further activation of Akt, which in turn inhibits GSK3β, suppressing tau phosphorylation [161]; (b) the LRP8-JIPS interaction at the plasma membrane level that interferes in turn with the signaling of JNK, whose translocation into the nucleus is therefore inhibited. In addition, the activity of rhoGEF, which is associated with JIP, might be modulated by the Reelin-LTP8-JIPs complex [18,162]; (c) the promotion of LRP8, Dab1 and NMDA-R clustering and related NMDA-R phosphorylation mediated by SFK, which in turn leads to an increase in calcium influx [163] (Chen, 2005). The signal activated by LRP8 and NMDA-R complex, but not necessarily their physical association, involves PSD-95, which is known to inhibit NMDA-R internalization [164].
Figure 3. LRP8 in CNS related molecular pathways and neurodegeneration. Ligands binding to LRP8 modulate intracellular pathways, LRP8 cleavage, LRP8-APP interaction and Aβ production. (1) ApoE. Aβ production is increased by ApoE binding to LRP8 [157]. (2) F-Spondin. F-Spondin increases LRP8 cleavage and decreases Aβ production [158]. (3) LRP8 levels also modulate Aβ production in a positive manner by increasing the APP association with lipid rafts and γ-secretase activity [159]. (4) Reelin. Reelin binding to LRP8 induces different signaling pathways: (a) the tyrosin phosphorylation of Dab1 by Src-family kinases [160] and its interaction with the LRP8 NPxY motif. Phosphorylated Dab1 subsequently interacts with PI3K, whose activation leads to the further activation of Akt, which in turn inhibits GSK3β, suppressing tau phosphorylation [161]; (b) the LRP8-JIPS interaction at the plasma membrane level that interferes in turn with the signaling of JNK, whose translocation into the nucleus is therefore inhibited. In addition, the activity of rhoGEF, which is associated with JIP, might be modulated by the Reelin-LTP8-JIPs complex [18,162]; (c) the promotion of LRP8, Dab1 and NMDA-R clustering and related NMDA-R phosphorylation mediated by SFK, which in turn leads to an increase in calcium influx [163] (Chen, 2005). The signal activated by LRP8 and NMDA-R complex, but not necessarily their physical association, involves PSD-95, which is known to inhibit NMDA-R internalization [164].
Ijms 23 08921 g003
LRP8 is mainly known for being a receptor of ApoE, which is produced in various organs, including liver, brain, kidneys, and adrenal glands, and which represents a component of lipoproteins responsible for packaging cholesterol and other fats and carrying them through the bloodstream [165]. LRP8 binds with high affinity only to ApoE-rich β-VLDL, while its affinity for LDL and other VLDL is much lower [120]. In humans, three ApoE isoforms exist that differ at positions 112 and 158 (ApoE2, ApoE3 and ApoE4) [166]. ApoE2 has cysteines at positions 112 and 158, ApoE3 has a cysteine at position 112 and an arginine at position 158 and ApoE4 has arginines at positions 112 and 158. The ApoE ε2 allele is associated with the recessive inheritance of hyperlipoproteinemia in patients with type III hyperlipidemia [167], and the ε4 allele is strongly associated with cardio-cerebrovascular diseases and Alzheimer’s disease [168]. A single copy of the ε4 allele increases AD risk by about 3-fold, with two copies of the ε4 allele increasing disease risk about 12-fold relative to individuals who have 2 copies of the ε3 allele. On the other side, the ε2 allele seems to decrease AD risk by about 0.6 [169,170]. Additional studies have demonstrated that ApoE is closely related to cognitive functions; indeed, ApoE-deficient homozygous mice show impaired working memory [171] accompanied by the age-dependent loss of synaptic proteins [172]. In addition to AD, ApoE has also been reported to have an impact on dementia and synucleinopathy in Parkinson’s disease (PD) dementia and dementia with Lewy bodies as well as in mouse models of synucleinopathy [173,174]. ApoE is also directly involved in the binding and clearance of Aβ, with ApoE ε3 and ApoE ε2 having a higher affinity compared with ApoE ε4 [175,176]. Moreover, these data were also confirmed in in vivo studies showing that knockout ApoE mice crossed with AD mice overexpressing APP exhibit less Aβ deposition compared with those expressing ApoE ε3 or ApoE ε4 [177,178].
In Alzheimer’s disease, Aβ is generated from APP through sequential cleavages, first by β-secretase and then by γ-secretase complex, the latter consisting of at least four components: presenilin (PS, PS1 or PS2), Nicastrin, anterior pharynx-defective-1 (APH-1) and presenilin enhancer-2 (PEN-2) [179,180]. PSs are the crucial catalytic components of γ-secretase, and mutations in PS1 are causative in the majority of familial AD (FAD) cases, likely through a yet-unclear loss of function, while APP overexpression or mutations are causative of FAD, likely favoring amyloidosis and tauopathy [181,182].
Hoe et al., uncovered interactions between LRP8, APP and F-spondin, a component of the extracellular matrix involved in neuronal migration and plasticity in adult and developing brains, suggesting that interaction between extracellular matrix proteins and LRP8 at the cell surface can alter APP processing and decrease Aβ production. LRP8 and APP interaction happens at the extracellular and intracellular levels and increases with the presence of F-spondin, which also mediates increased LRP8 cleavage [158].
The potential clustering of APP and LRP8 modulates their endocytosis, likely altering the cleavage of each receptor, via both extracellular metalloproteinases and intramembranous γ-secretase cleavage. Interestingly, LRP8 expression also correlates with a significant increase in Aβ production and reduced levels of APP-CTFs. The increased Aβ production seems, in turn, to depend on the integrity of the NPXY endocytosis motif of LRP8; in fact, LRP8 expression increases APP association with lipid rafts and γ-secretase activity, both of which might contribute to increased Aβ production [159]. Moreover, ApoE binding to LRP8 also increases APP endocytosis and Aβ production in an isoform-specific manner [157]. There is therefore a link between ApoE and Aβ that does not depend on their direct interaction but rather on the ability of the different isoforms of ApoE to modify the processing of APP through LRP8, thus representing a bidirectional bridge between cholesterol metabolism and amyloidosis.
In addition to ApoE, another well-known ligand of LRP8 is Reelin: which is ligand as well of the VLDL receptor and that is produced by Cajal–Retzius neurons at the surface of the developing neocortex [183]. Depending on the specific tissue and cell type involved, the binding of Reelin to LRP8 leads to the tyrosine phosphorylation of the adaptor protein Dab1/2 to initiate several signaling cascades such as the activation of PI3K, ERK1/2, Src-family kinases and protein kinase B/Akt. In this context, it is important to highlight that Dab1 is essential for neuronal migration both upon LRP8 or APP activation, suggesting that it is likely through Dab1 that both APP and LRP8 regulate neuronal migration from SVZ toward the cortical plate [59,184]. Furthermore, Stockinger et al., have reported a role of the LRP8-Reelin interaction in the correct positioning of neurons during the embryonic development of the brain.
In the CNS, Reelin is able to regulate APP processing and Aβ production by interacting with APP. Reelin increases cell surface levels of APP and decreasedthe endocytosis of APP in hippocampal neurons in vitro. In vivo, Reelin levels were increased in the brains of APP knockout mice and decreased in APP-overexpressing mice [185].
Reelin signaling is crucial for many CNS functions such as neuronal migration, dendritic spine development [186], synaptic plasticity [184,187,188] and brain embryonic development [59]. Some of the Reelin functions are played through binding to LRP8; indeed, LRP8 Reelin-regulated neuronal (LRN) enhancer binding modulates learning and memory, which activates synaptic plasticity genes including NMDA receptor NR1, NR2A and NR2B [124]. In addition to its NMDA gene activation, Reelin signaling through LRP8 induces the tyrosine phosphorylation of the receptor NMDA (NR2) subunits, changing their surface distribution and activity and in turn regulating synaptic plasticity, which is further modulated by the differential splicing of the LRP8 cytoplasmic domain [188]. Moreover, LRP8 proline rich 59 amino acid insert is essential for the Reelin-induced enhancement of LTP in mice [59]; in this context, interference with Reelin expression or its intracellular signaling pathway has effects on LTP and seizure potential that are partially replicated in LRP8 knockouts. In this context, presenilin mutations have also been linked to Reelin-signaling elements such as phosphatidylinositide 3-kinase and Crk/Dock1/Rac, highlighting a potential correlation of presenilin mutations on synaptic processes with LRP8 [189,190].
Additional studies on LRP8-KO mice have supported a role of LRP8 in neuronal functions, showing in particular a severe impairment in freezing behaviors by performing a classical form of robust associative learning (contextual fear paradigm) reflecting a decline in long-term memory formation [124]. Moreover, LRP8 deficiency in mice has been shown to affect the development of the neocortex, preventing neurons from completing their migration and causing in turn a partial inversion of the neuronal layers in the neocortex [59]. In this context, LRP8 KO/ApoE KO mice have shown cognitive decline greater than that observed in ApoE single-KO mice as indicated by behavioral tests [191].
In addition to the activation of intracellular signaling pathways, the binding of some ligands, especially Reelin, to LRP8 also regulates its proteolytic processing. From the LRP8 full-length mature form (105 kDa), two major proteolytic products have been described so far [88,166,167,168], the carboxy-terminal fragment generated by extracellular α-secretase (20–25 kDa) and a C-terminal fragment (15–18 kDa) that is produced by γ-secretase cleavage and is similar to AICDs derived by APP processing [166]. In fact, LRP8 is a γ-secretase substrate, like APP and other LDL members such as LRP1 [169], LRP1b [170], LRP2 [171], LRP6 [172], LR11 [173] and VLDLR [174] as well as many other substrates including Notch [175,176,177,178]. The LRP8 C-terminal fragment produced by γ-secretase cleavage appears to translocate into the nucleus where it inhibits Reelin transcription, creating a negative feedback loop for Reelin levels; indeed, depletion of Presenilin1, the catalytic component of γ-secretase, elevates the levels of both Reelin and LRP8 in mouse brain [123]. Telese et al., showed that the nuclear translocation of the LRP8 CTF, derived from γ-secretase cut, stimulates specific enhancers critically involved in hippocampus-dependent learning and memory, implying that Reelin can impact genes related to learning and memory via the LRP8 γ-CTF and that this process might be attenuated by PS1 mutations [124]. In this scenario, in which the processing of LRP8 modulates its functions, Reelin causes a reduction of the full-length receptor, probably enhancing its proteolysis by a sheddase after ligand-stimulated internalization. PS1 mutations show impaired γ-secretase activity relatively to LRP8 and an accumulation of its C-terminal fragments after Reelin exposure; this last effect is potentiated by co-exposure to DAPT, a γ-secretase inhibitor [192].
In addition to the ligands already mentioned, ApoE, F-Spondin and Reelin, other proposed LRP8 ligands are: (1) trombospondin-1, expressed in the subventricular zone and throughout the rostral migratory stream, where it acts on LRP8 by promoting neuroblast chain migration [193]; (2) clusterin (apolipoprotein J), which modulates a cell proliferative signal in migrating neuronal precursors of SVZ explants, in vitro, via Dab1 activation [194]; (3) selenoprotein P, a mediator of selenium transport whose binding with LRP8 has been observed in the brain, testis and bone. Selenium and its carrier protein, selenoprotein P, have been reported to have important roles in maintaining optimal brain function as well as in relevant AD-related pathways in addition to regulating the development and the immune system and having antitumor properties due to their strong antioxidant activities [195]. Epidemiological studies have shown a significant positive correlation between selenium level and cognitive ability [196] and that blood selenium level gradually decreases with age [197]. Furthermore, selenium levels change significantly in the brain and blood of patients with various neurodegenerative diseases such as AD, PD, multiple sclerosis and Batten’s disease [198]. In this context, LRP8 KO mice have shown similar phenotypes of selenoprotein P KO mice in the brain and in the testis, highlighting the biological significance of the receptor-mediated uptake of selenium. Indeed, lowering brain Se levels by the genetic inactivation of selenoprotein P, or its neuronal receptor LRP8 [199,200], has been shown to lead to spontaneous neurological deficits and neurodegeneration.
An overview of the LRP8 roles and molecular mechanisms in CNS and neurodegeneration is reported in Table 3.

4. Final Remarks

LDLR, and LRP8 in particular, are receptors that through multiple pathways and mechanisms seems to play a role both in the modulation of neuronal activity and in the regulation of cell proliferation. In this context, several studies have proposed a potential link between oncological and neurodegenerative pathways but have rarely found a common denominator. Using an obvious oversimplification, we could say that high levels of LRP8 manifest a correlation with proliferation and metastasis in cancer cells and tissues, while functions related to neuronal migration, amyloidosis and neurodegeneration, in a broad sense, are likely modulated by the proteolytic processing of LRP8, which however has never been fully defined in its complexity.
Some features of LRP8 are, from this point of view, relevant: from one side, the same pathways (for example Wnt) foresee LRP8 as pivotal regulator both in degeneration like Alzheimer’s and in tumor genesis such as in breast cancer; from the other side, ApoE, the most famous ligand of LRP8, is at the crossroads between metastatic proliferation and Alzheimer’s-type neurodegeneration. At present, it is not clear what is the main pathway through which LRP8 can influence tumor proliferation or whether there is a prevalent mechanism in which it is involved. Specific studies on cancer stem cells and using in vivo xenograft animal models will need to clarify these aspects before LRP8 can be defined as a potential therapeutic marker.
In addition, it is well-known that the processing of many receptors involved in oncological and/or neurodegenerative pathways depends on the action of the intramembrane protease γ-secretase. In fact, γ-secretase inhibitors are still used in oncological trials and in the recent past have been used, with unfortunately negative results, to treat patients suffering from Alzheimer’s disease. Regarding LRP8, it is not known if its processing by γ-secretase is altered in patients with Alzheimer’s disease, and the consequences of this processing on amyloid formation are still underexplored.
After putting together the pieces of the puzzle, there are still obvious questions that remain open and whose answers could lead not only to decipher key points in the intersection between oncological and neurodegenerative pathways but also to define new therapeutic targets in both fields: One, what is the exact role of ApoE/LRP8 in tumorigenesis and Alzheimer’s? Two, how important are the different isoforms of splicing and the complex processing of LRP8 to defining its functions in AD and cancer? Three, can amyloidosis and cell proliferation in cancer be reversed targeting ApoE/LRP8? In this context, it is sadly remarkable that the hypothesis of a loss of function of γ-secretase in familial AD mutants, relative to other substrates than APP, has never been studied in depth [201,202]. The answers to these questions will hopefully lead to a greater knowledge of the molecular mechanisms in which LDLR receptors are involved, and LRP8 in particular, to develop new diagnostic and therapeutic tools that can be used both in oncology and in some neurodegenerative conditions such as the Alzheimer’s dementia and pathologies related to aberrant neuronal migration.

Author Contributions

Conceptualization, C.P., D.P. and C.R.; Bibliographic investigation, V.D.L., A.M., E.F. and M.F.; data curation, A.M., D.M., M.R. and M.F.; writing—original draft preparation, D.P. and C.P.; writing—review and editing, D.P., S.C., V.D.L. and M.Z.; supervision, S.C. and C.P.; funding acquisition, C.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by “Scientific projects referred to in art. 1 paragraph 34 and 34 bis, L. 12.23.1996, n. 662 for the implementation of the objectives of a priority nature and of national importance (D15I13000430001)” to C.R.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors wish to thank Francesco Caciagli and Patrizia di Iorio for their tireless scientific and knowledge support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Karanth, S.D.; Katsumata, Y.; Nelson, P.T.; Fardo, D.W.; McDowell, J.K.; Schmitt, F.A.; Kryscio, R.J.; Browning, S.R.; Braithwaite, D.; Arnold, S.M.; et al. Cancer Diagnosis Is Associated with a Lower Burden of Dementia and Less Alzheimer’s-Type Neuropathology. Brain 2022, 145, 2518–2527. [Google Scholar] [CrossRef] [PubMed]
  2. Ospina-Romero, M.; Glymour, M.M.; Hayes-Larson, E.; Mayeda, E.R.; Graff, R.E.; Brenowitz, W.D.; Ackley, S.F.; Witte, J.S.; Kobayashi, L.C. Association Between Alzheimer Disease and Cancer with Evaluation of Study Biases. JAMA Netw. Open 2020, 3, e2025515. [Google Scholar] [CrossRef]
  3. Driver, J.A. Understanding the Link between Cancer and Neurodegeneration. J. Geriatr. Oncol. 2012, 3, 58–67. [Google Scholar] [CrossRef]
  4. Li, Y.; Cam, J.; Bu, G. Low-Density Lipoprotein Receptor Family. Mol. Neurobiol. 2001, 23, 53–67. [Google Scholar] [CrossRef]
  5. Lane-Donovan, C.E.; Philips, G.T.; Herz, J. More Than Cholesterol Transporters: Lipoprotein Receptors in CNS Function and Neurodegeneration. Neuron 2014, 83, 771–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Roslan, Z.; Muhamad, M.; Selvaratnam, L.; Ab-Rahim, S. The Roles of Low-Density Lipoprotein Receptor-Related Proteins 5, 6, and 8 in Cancer: A Review. J. Oncol. 2019, 2019, e4536302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Andersen, R.K.; Hammer, K.; Hager, H.; Christensen, J.N.; Ludvigsen, M.; Honoré, B.; Thomsen, M.-B.H.; Madsen, M. Melanoma Tumors Frequently Acquire LRP2/Megalin Expression, Which Modulates Melanoma Cell Proliferation and Survival Rates. Pigment. Cell Melanoma Res. 2015, 28, 267–280. [Google Scholar] [CrossRef]
  8. Yang, P.; Liu, Z.; Wang, H.; Tian, J.; Li, Y.; Zong, Y.; Qu, S. Enhanced Activity of Very Low Density Lipoprotein Receptor II Promotes SGC7901 Cell Proliferation and Migration. Life Sci. 2009, 84, 402–408. [Google Scholar] [CrossRef]
  9. Lin, C.-C.; Lo, M.-C.; Moody, R.; Jiang, H.; Harouaka, R.; Stevers, N.; Tinsley, S.; Gasparyan, M.; Wicha, M.; Sun, D. Targeting LRP8 Inhibits Breast Cancer Stem Cells in Triple-Negative Breast Cancer. Cancer Lett. 2018, 438, 165–173. [Google Scholar] [CrossRef]
  10. Gent, J.; Braakman, I. Low-Density Lipoprotein Receptor Structure and Folding. CMLS Cell. Mol. Life Sci. 2004, 61, 2461–2470. [Google Scholar] [CrossRef]
  11. Andersen, O.M.; Dagil, R.; Kragelund, B.B. New Horizons for Lipoprotein Receptors: Communication by β-Propellers. J. Lipid. Res. 2013, 54, 2763–2774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Dlugosz, P.; Nimpf, J. The Reelin Receptors Apolipoprotein E Receptor 2 (ApoER2) and VLDL Receptor. Int. J. Mol. Sci. 2018, 19, 3090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Borg, J.P.; Ooi, J.; Levy, E.; Margolis, B. The Phosphotyrosine Interaction Domains of X11 and FE65 Bind to Distinct Sites on the YENPTY Motif of Amyloid Precursor Protein. Mol. Cell Biol. 1996, 16, 6229–6241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Batzer, A.G.; Blaikie, P.; Nelson, K.; Schlessinger, J.; Margolis, B. The Phosphotyrosine Interaction Domain of Shc Binds an LXNPXY Motif on the Epidermal Growth Factor Receptor. Mol. Cell Biol. 1995, 15, 4403–4409. [Google Scholar] [CrossRef] [Green Version]
  15. Calderwood, D.A.; Yan, B.; de Pereda, J.M.; Alvarez, B.G.; Fujioka, Y.; Liddington, R.C.; Ginsberg, M.H. The Phosphotyrosine Binding-like Domain of Talin Activates Integrins. J. Biol. Chem. 2002, 277, 21749–21758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Gotthardt, M.; Trommsdorff, M.; Nevitt, M.F.; Shelton, J.; Richardson, J.A.; Stockinger, W.; Nimpf, J.; Herz, J. Interactions of the Low Density Lipoprotein Receptor Gene Family with Cytosolic Adaptor and Scaffold Proteins Suggest Diverse Biological Functions in Cellular Communication and Signal Transduction. J. Biol. Chem. 2000, 275, 25616–25624. [Google Scholar] [CrossRef] [Green Version]
  17. Trommsdorff, M.; Borg, J.-P.; Margolis, B.; Herz, J. Interaction of Cytosolic Adaptor Proteins with Neuronal Apolipoprotein E Receptors and the Amyloid Precursor Protein. J. Biol. Chem. 1998, 273, 33556–33560. [Google Scholar] [CrossRef] [Green Version]
  18. Stockinger, W.; Brandes, C.; Fasching, D.; Hermann, M.; Gotthardt, M.; Herz, J.; Schneider, W.J.; Nimpf, J. The Reelin Receptor ApoER2 Recruits JNK-Interacting Proteins-1 and -2. J. Biol. Chem. 2000, 275, 25625–25632. [Google Scholar] [CrossRef] [Green Version]
  19. Go, G.; Mani, A. Low-Density Lipoprotein Receptor (LDLR) Family Orchestrates Cholesterol Homeostasis. Yale J. Biol. Med. 2012, 85, 19–28. [Google Scholar]
  20. Weisgraber, K.H. Apolipoprotein E: Structure-Function Relationships. Adv. Protein. Chem. 1994, 45, 249–302. [Google Scholar] [CrossRef]
  21. Boyles, J.K.; Pitas, R.E.; Wilson, E.; Mahley, R.W.; Taylor, J.M. Apolipoprotein E Associated with Astrocytic Glia of the Central Nervous System and with Nonmyelinating Glia of the Peripheral Nervous System. J. Clin. Investig. 1985, 76, 1501–1513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Mahley, R.W. Apolipoprotein E: Cholesterol Transport Protein with Expanding Role in Cell Biology. Science 1988, 240, 622–630. [Google Scholar] [CrossRef]
  23. Johnson, L.A.; Olsen, R.H.J.; Merkens, L.S.; DeBarber, A.; Steiner, R.D.; Sullivan, P.M.; Maeda, N.; Raber, J. Apolipoprotein E–Low Density Lipoprotein Receptor Interaction Affects Spatial Memory Retention and Brain ApoE Levels in an Isoform-Dependent Manner. Neurobiol. Dis. 2014, 64, 150–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Abisambra, J.F.; Fiorelli, T.; Padmanabhan, J.; Neame, P.; Wefes, I.; Potter, H. LDLR Expression and Localization Are Altered in Mouse and Human Cell Culture Models of Alzheimer’s Disease. PLoS ONE 2010, 5, e8556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Katsouri, L.; Georgopoulos, S. Lack of LDL Receptor Enhances Amyloid Deposition and Decreases Glial Response in an Alzheimer’s Disease Mouse Model. PLoS ONE 2011, 6, e21880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Fryer, J.D.; Demattos, R.B.; McCormick, L.M.; O’Dell, M.A.; Spinner, M.L.; Bales, K.R.; Paul, S.M.; Sullivan, P.M.; Parsadanian, M.; Bu, G.; et al. The Low Density Lipoprotein Receptor Regulates the Level of Central Nervous System Human and Murine Apolipoprotein E but Does Not Modify Amyloid Plaque Pathology in PDAPP Mice. J. Biol. Chem 2005, 280, 25754–25759. [Google Scholar] [CrossRef] [Green Version]
  27. Kim, J.; Castellano, J.M.; Jiang, H.; Basak, J.M.; Parsadanian, M.; Pham, V.; Mason, S.M.; Paul, S.M.; Holtzman, D.M. Overexpression of Low-Density Lipoprotein Receptor in the Brain Markedly Inhibits Amyloid Deposition and Increases Extracellular A Beta Clearance. Neuron 2009, 64, 632–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. 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] [CrossRef] [PubMed] [Green Version]
  29. Chen, Z.; Chen, L.; Sun, B.; Liu, D.; He, Y.; Qi, L.; Li, G.; Han, Z.; Zhan, L.; Zhang, S.; et al. LDLR Inhibition Promotes Hepatocellular Carcinoma Proliferation and Metastasis by Elevating Intracellular Cholesterol Synthesis through the MEK/ERK Signaling Pathway. Mol. Metab. 2021, 51, 101230. [Google Scholar] [CrossRef]
  30. Lin, L.; Hu, K. LRP-1: Functions, Signaling and Implications in Kidney and Other Diseases. Int. J. Mol. Sci. 2014, 15, 22887–22901. [Google Scholar] [CrossRef] [Green Version]
  31. Zlokovic, B.V.; Deane, R.; Sagare, A.P.; Bell, R.D.; Winkler, E.A. Low-Density Lipoprotein Receptor-Related Protein-1: A Serial Clearance Homeostatic Mechanism Controlling Alzheimer’s Amyloid β-Peptide Elimination from the Brain. J. Neurochem. 2010, 115, 1077–1089. [Google Scholar] [CrossRef] [Green Version]
  32. Sagare, A.P.; Deane, R.; Zlokovic, B.V. Low-Density Lipoprotein Receptor-Related Protein 1: A Physiological Aβ Homeostatic Mechanism with Multiple Therapeutic Opportunities. Pharmacol. Ther. 2012, 136, 94–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kanekiyo, T.; Bu, G. The Low-Density Lipoprotein Receptor-Related Protein 1 and Amyloid-β Clearance in Alzheimer’s Disease. Front. Aging Neurosci. 2014, 6, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Qiu, Z.; Crutcher, K.A.; Hyman, B.T.; Rebeck, G.W. ApoE Isoforms Affect Neuronal N-Methyl-D-Aspartate Calcium Responses and Toxicity via Receptor-Mediated Processes. Neuroscience 2003, 122, 291–303. [Google Scholar] [CrossRef] [PubMed]
  35. Hayashi, H.; Campenot, R.B.; Vance, D.E.; Vance, J.E. Apolipoprotein E-Containing Lipoproteins Protect Neurons from Apoptosis via a Signaling Pathway Involving Low-Density Lipoprotein Receptor-Related Protein-1. J. Neurosci. 2007, 27, 1933–1941. [Google Scholar] [CrossRef]
  36. Ulery, P.G.; Beers, J.; Mikhailenko, I.; Tanzi, R.E.; Rebeck, G.W.; Hyman, B.T.; Strickland, D.K. Modulation of Beta-Amyloid Precursor Protein Processing by the Low Density Lipoprotein Receptor-Related Protein (LRP). Evidence That LRP Contributes to the Pathogenesis of Alzheimer’s Disease. J. Biol. Chem. 2000, 275, 7410–7415. [Google Scholar] [CrossRef] [Green Version]
  37. Pietrzik, C.U. The Cytoplasmic Domain of the LDL Receptor-Related Protein Regulates Multiple Steps in APP Processing. EMBO J. 2002, 21, 5691–5700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Ramanathan, A.; Nelson, A.R.; Sagare, A.P.; Zlokovic, B.V. Impaired Vascular-Mediated Clearance of Brain Amyloid Beta in Alzheimer’s Disease: The Role, Regulation and Restoration of LRP1. Front. Aging Neurosci. 2015, 7, 136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Yepes, M.; Sandkvist, M.; Moore, E.G.; Bugge, T.H.; Strickland, D.K.; Lawrence, D.A. Tissue-Type Plasminogen Activator Induces Opening of the Blood-Brain Barrier via the LDL Receptor–Related Protein. J. Clin. Investig. 2003, 112, 1533–1540. [Google Scholar] [CrossRef] [PubMed]
  40. Herz, J.; Clouthier, D.E.; Hammer, R.E. LDL Receptor-Related Protein Internalizes and Degrades UPA-PAI-1 Complexes and Is Essential for Embryo Implantation. Cell 1992, 71, 411–421. [Google Scholar] [CrossRef]
  41. May, P.; Rohlmann, A.; Bock, H.H.; Zurhove, K.; Marth, J.D.; Schomburg, E.D.; Noebels, J.L.; Beffert, U.; Sweatt, J.D.; Weeber, E.J.; et al. Neuronal LRP1 Functionally Associates with Postsynaptic Proteins and Is Required for Normal Motor Function in Mice. Mol. Cell Biol. 2004, 24, 8872–8883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. 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] [CrossRef] [PubMed] [Green Version]
  43. Song, H.; Li, Y.; Lee, J.; Schwartz, A.L.; Bu, G. Low-Density Lipoprotein Receptor-Related Protein 1 Promotes Cancer Cell Migration and Invasion by Inducing the Expression of Matrix Metalloproteinases 2 and 9. Cancer Res. 2009, 69, 879–886. [Google Scholar] [CrossRef] [Green Version]
  44. Fayard, B.; Bianchi, F.; Dey, J.; Moreno, E.; Djaffer, S.; Hynes, N.E.; Monard, D. The Serine Protease Inhibitor Protease Nexin-1 Controls Mammary Cancer Metastasis through LRP-1–Mediated MMP-9 Expression. Cancer Res. 2009, 69, 5690–5698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Catasus, L.; Gallardo, A.; Llorente-Cortes, V.; Escuin, D.; Muñoz, J.; Tibau, A.; Peiro, G.; Barnadas, A.; Lerma, E. Low-Density Lipoprotein Receptor–Related Protein 1 Is Associated with Proliferation and Invasiveness in Her-2/Neu and Triple-Negative Breast Carcinomas. Hum. Pathol. 2011, 42, 1581–1588. [Google Scholar] [CrossRef] [PubMed]
  46. Christensen, E.I.; Birn, H.; Storm, T.; Weyer, K.; Nielsen, R. Endocytic Receptors in the Renal Proximal Tubule. Physiology 2012, 27, 223–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Carro, E.; Spuch, C.; Trejo, J.L.; Antequera, D.; Torres-Aleman, I. Choroid Plexus Megalin Is Involved in Neuroprotection by Serum Insulin-Like Growth Factor I. J. Neurosci. 2005, 25, 10884–10893. [Google Scholar] [CrossRef]
  48. Gajera, C.R.; Emich, H.; Lioubinski, O.; Christ, A.; Beckervordersandforth-Bonk, R.; Yoshikawa, K.; Bachmann, S.; Christensen, E.I.; Götz, M.; Kempermann, G.; et al. LRP2 in Ependymal Cells Regulates BMP Signaling in the Adult Neurogenic Niche. J. Cell Sci. 2010, 123, 1922–1930. [Google Scholar] [CrossRef] [Green Version]
  49. Gomes, J.R.; Lobo, A.; Nogueira, R.; Terceiro, A.F.; Costelha, S.; Lopes, I.M.; Magalhães, A.; Summavielle, T.; Saraiva, M.J. Neuronal Megalin Mediates Synaptic Plasticity-a Novel Mechanism Underlying Intellectual Disabilities in Megalin Gene Pathologies. Brain Commun. 2020, 2, fcaa135. [Google Scholar] [CrossRef] [PubMed]
  50. Marzolo, M.-P.; Farfán, P. New Insights into the Roles of Megalin/LRP2 and the Regulation of Its Functional Expression. Biol. Res. 2011, 44, 89–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Gomes, J.R.; Sárkány, Z.; Teixeira, A.; Nogueira, R.; Cabrito, I.; Soares, H.; Wittelsberger, A.; Stortelers, C.; Macedo-Ribeiro, S.; Vanlandschoot, P.; et al. Anti-TTR Nanobodies Allow the Identification of TTR Neuritogenic Epitope Associated with TTR-Megalin Neurotrophic Activities. ACS Chem. Neurosci. 2019, 10, 704–715. [Google Scholar] [CrossRef]
  52. Fitzgerald, M.; Nairn, P.; Bartlett, C.A.; Chung, R.S.; West, A.K.; Beazley, L.D. Metallothionein-IIA Promotes Neurite Growth via the Megalin Receptor. Exp. Brain Res. 2007, 183, 171–180. [Google Scholar] [CrossRef] [PubMed]
  53. Alvira-Botero, X.; Pérez-Gonzalez, R.; Spuch, C.; Vargas, T.; Antequera, D.; Garzón, M.; Bermejo-Pareja, F.; Carro, E. Megalin Interacts with APP and the Intracellular Adapter Protein FE65 in Neurons. Mol. Cell. Neurosci. 2010, 45, 306–315. [Google Scholar] [CrossRef] [PubMed]
  54. Chiu-Ugalde, J.; Theilig, F.; Behrends, T.; Drebes, J.; Sieland, C.; Subbarayal, P.; Köhrle, J.; Hammes, A.; Schomburg, L.; Schweizer, U. Mutation of Megalin Leads to Urinary Loss of Selenoprotein P and Selenium Deficiency in Serum, Liver, Kidneys and Brain. Biochem. J. 2010, 431, 103–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Pober, B.R.; Longoni, M.; Noonan, K.M. A Review of Donnai-Barrow and Facio-Oculo-Acoustico-Renal (DB/FOAR) Syndrome: Clinical Features and Differential Diagnosis. Birth Defects Res. Part. A Clin. Mol. Teratol. 2009, 85, 76–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Vargas, T.; Bullido, M.J.; Martinez-Garcia, A.; Antequera, D.; Clarimon, J.; Rosich-Estrago, M.; Martin-Requero, A.; Mateo, I.; Rodriguez-Rodriguez, E.; Vilella-Cuadrada, E.; et al. A Megalin Polymorphism Associated with Promoter Activity and Alzheimer’s Disease Risk. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2010, 153B, 895–902. [Google Scholar] [CrossRef]
  57. Holt, S.K.; Karyadi, D.M.; Kwon, E.M.; Stanford, J.L.; Nelson, P.S.; Ostrander, E.A. Association of Megalin Genetic Polymorphisms with Prostate Cancer Risk and Prognosis. Clin. Cancer Res. 2008, 14, 3823–3831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Ge, S.; Li, B.; Li, Y.; Li, Z.; Liu, Z.; Chen, Z.; Wu, J.; Gao, J.; Shen, L. Genomic Alterations in Advanced Gastric Cancer Endoscopic Biopsy Samples Using Targeted Next-Generation Sequencing. Am. J. Cancer Res. 2017, 7, 1540–1553. [Google Scholar]
  59. Trommsdorff, M.; Gotthardt, M.; Hiesberger, T.; Shelton, J.; Stockinger, W.; Nimpf, J.; Hammer, R.E.; Richardson, J.A.; Herz, J. Reeler/Disabled-like Disruption of Neuronal Migration in Knockout Mice Lacking the VLDL Receptor and ApoE Receptor 2. Cell 1999, 97, 689–701. [Google Scholar] [CrossRef] [Green Version]
  60. Zhang, G.; Assadi, A.H.; McNeil, R.S.; Beffert, U.; Wynshaw-Boris, A.; Herz, J.; Clark, G.D.; D’Arcangelo, G. The Pafah1b Complex Interacts with the Reelin Receptor VLDLR. PLoS ONE 2007, 2, e252. [Google Scholar] [CrossRef] [PubMed]
  61. He, L.; Lu, Y.; Wang, P.; Zhang, J.; Yin, C.; Qu, S. Up-Regulated Expression of Type II Very Low Density Lipoprotein Receptor Correlates with Cancer Metastasis and Has a Potential Link to β-Catenin in Different Cancers. BMC Cancer 2010, 10, 601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Leupin, O.; Piters, E.; Halleux, C.; Hu, S.; Kramer, I.; Morvan, F.; Bouwmeester, T.; Schirle, M.; Bueno-Lozano, M.; Fuentes, F.J.R.; et al. Bone Overgrowth-Associated Mutations in the LRP4 Gene Impair Sclerostin Facilitator Function. J. Biol. Chem. 2011, 286, 19489–19500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Sun, X.-D.; Li, L.; Liu, F.; Huang, Z.-H.; Bean, J.C.; Jiao, H.-F.; Barik, A.; Kim, S.-M.; Wu, H.; Shen, C.; et al. Lrp4 in Astrocytes Modulates Glutamatergic Transmission. Nat. Neurosci. 2016, 19, 1010–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhang, H.; Chen, W.; Tan, Z.; Zhang, L.; Dong, Z.; Cui, W.; Zhao, K.; Wang, H.; Jing, H.; Cao, R.; et al. A Role of Low-Density Lipoprotein Receptor-Related Protein 4 (LRP4) in Astrocytic Aβ Clearance. J. Neurosci. 2020, 40, 5347–5361. [Google Scholar] [CrossRef]
  65. Gomez, A.M.; Froemke, R.C.; Burden, S.J. Synaptic Plasticity and Cognitive Function Are Disrupted in the Absence of Lrp4. eLife 2014, 3, e04287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zhang, H.; Sathyamurthy, A.; Liu, F.; Li, L.; Zhang, L.; Dong, Z.; Cui, W.; Sun, X.; Zhao, K.; Wang, H.; et al. Agrin-Lrp4-Ror2 Signaling Regulates Adult Hippocampal Neurogenesis in Mice. eLife 2019, 8, e45303. [Google Scholar] [CrossRef] [PubMed]
  67. Kim, N.; Stiegler, A.L.; Cameron, T.O.; Hallock, P.T.; Gomez, A.M.; Huang, J.H.; Hubbard, S.R.; Dustin, M.L.; Burden, S.J. Lrp4 Is a Receptor for Agrin and Forms a Complex with MuSK. Cell 2008, 135, 334–342. [Google Scholar] [CrossRef] [Green Version]
  68. Zhang, B.; Luo, S.; Wang, Q.; Suzuki, T.; Xiong, W.C.; Mei, L. LRP4 Serves as a Coreceptor of Agrin. Neuron 2008, 60, 285–297. [Google Scholar] [CrossRef] [Green Version]
  69. Zong, Y.; Zhang, B.; Gu, S.; Lee, K.; Zhou, J.; Yao, G.; Figueiredo, D.; Perry, K.; Mei, L.; Jin, R. Structural Basis of Agrin–LRP4–MuSK Signaling. Genes Dev. 2012, 26, 247–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Zhou, X.; Xia, E.; Bhandari, A.; Zheng, C.; Xiang, J.; Guan, Y.; Zhang, X. LRP4 Promotes Proliferation, Migration, and Invasion in Papillary Thyroid Cancer. Biochem. Biophys. Res. Commun. 2018, 503, 257–263. [Google Scholar] [CrossRef] [PubMed]
  71. Mao, Z.; Wang, Z.; Zhang, S.; Pu, Y.; Wang, J.; Zhang, T.; Long, Y.; Liu, Y.; Ma, Y.; Zhu, J. LRP4 Promotes Migration and Invasion of Gastric Cancer under the Regulation of MicroRNA-140-5p. CBM 2020, 29, 245–253. [Google Scholar] [CrossRef] [PubMed]
  72. Offe, K.; Dodson, S.E.; Shoemaker, J.T.; Fritz, J.J.; Gearing, M.; Levey, A.I.; Lah, J.J. The Lipoprotein Receptor LR11 Regulates Amyloid Beta Production and Amyloid Precursor Protein Traffic in Endosomal Compartments. J. Neurosci. 2006, 26, 1596–1603. [Google Scholar] [CrossRef] [Green Version]
  73. Andersen, O.M.; Reiche, J.; Schmidt, V.; Gotthardt, M.; Spoelgen, R.; Behlke, J.; von Arnim, C.A.F.; Breiderhoff, T.; Jansen, P.; Wu, X.; et al. Neuronal Sorting Protein-Related Receptor SorLA/LR11 Regulates Processing of the Amyloid Precursor Protein. Proc. Natl. Acad. Sci. USA 2005, 102, 13461–13466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Schmidt, V.; Sporbert, A.; Rohe, M.; Reimer, T.; Rehm, A.; Andersen, O.M.; Willnow, T.E. SorLA/LR11 Regulates Processing of Amyloid Precursor Protein via Interaction with Adaptors GGA and PACS-1*. J. Biol. Chem. 2007, 282, 32956–32964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ciarlo, E.; Massone, S.; Penna, I.; Nizzari, M.; Gigoni, A.; Dieci, G.; Russo, C.; Florio, T.; Cancedda, R.; Pagano, A. An Intronic NcRNA-Dependent Regulation of SORL1 Expression Affecting Aβ Formation Is Upregulated in Post-Mortem Alzheimer’s Disease Brain Samples. Dis. Model. Mech. 2013, 6, 424–433. [Google Scholar] [CrossRef] [Green Version]
  76. Scherzer, C.R.; Offe, K.; Gearing, M.; Rees, H.D.; Fang, G.; Heilman, C.J.; Schaller, C.; Bujo, H.; Levey, A.I.; Lah, J.J. Loss of Apolipoprotein E Receptor LR11 in Alzheimer Disease. Arch. Neurol. 2004, 61, 1200–1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Rogaeva, E.; Meng, Y.; Lee, J.H.; Gu, Y.; Kawarai, T.; Zou, F.; Katayama, T.; Baldwin, C.T.; Cheng, R.; Hasegawa, H.; et al. The Neuronal Sortilin-Related Receptor SORL1 Is Genetically Associated with Alzheimer Disease. Nat. Genet. 2007, 39, 168–177. [Google Scholar] [CrossRef] [PubMed]
  78. Pietilä, M.; Sahgal, P.; Peuhu, E.; Jäntti, N.Z.; Paatero, I.; Närvä, E.; Al-Akhrass, H.; Lilja, J.; Georgiadou, M.; Andersen, O.M.; et al. SORLA Regulates Endosomal Trafficking and Oncogenic Fitness of HER2. Nat. Commun. 2019, 10, 2340. [Google Scholar] [CrossRef] [Green Version]
  79. Sakai, S.; Nakaseko, C.; Takeuchi, M.; Ohwada, C.; Shimizu, N.; Tsukamoto, S.; Kawaguchi, T.; Jiang, M.; Sato, Y.; Ebinuma, H.; et al. Circulating Soluble LR11/SorLA Levels Are Highly Increased and Ameliorated by Chemotherapy in Acute Leukemias. Clinica. Chimica. Acta 2012, 413, 1542–1548. [Google Scholar] [CrossRef] [PubMed]
  80. Kawaguchi, T.; Ohwada, C.; Takeuchi, M.; Shimizu, N.; Sakaida, E.; Takeda, Y.; Sakai, S.; Tsukamoto, S.; Yamazaki, A.; Sugita, Y.; et al. LR11: A Novel Biomarker Identified in Follicular Lymphoma. Br. J. Haematol. 2013, 163, 277–280. [Google Scholar] [CrossRef] [PubMed]
  81. Brown, S.D.; Twells, R.C.J.; Hey, P.J.; Cox, R.D.; Levy, E.R.; Soderman, A.R.; Metzker, M.L.; Caskey, C.T.; Todd, J.A.; Hess, J.F. Isolation and Characterization OfLRP6,a Novel Member of the Low Density Lipoprotein Receptor Gene Family. Biochem. Biophys. Res. Commun. 1998, 248, 879–888. [Google Scholar] [CrossRef]
  82. DeBruine, Z.J.; Xu, H.E.; Melcher, K. Assembly and Architecture of the Wnt/β-Catenin Signalosome at the Membrane. Br. J. Pharmacol. 2017, 174, 4564–4574. [Google Scholar] [CrossRef]
  83. Pinson, K.I.; Brennan, J.; Monkley, S.; Avery, B.J.; Skarnes, W.C. An LDL-Receptor-Related Protein Mediates Wnt Signalling in Mice. Nature 2000, 407, 535–538. [Google Scholar] [CrossRef] [PubMed]
  84. Tamai, K.; Semenov, M.; Kato, Y.; Spokony, R.; Liu, C.; Katsuyama, Y.; Hess, F.; Saint-Jeannet, J.P.; He, X. LDL-Receptor-Related Proteins in Wnt Signal Transduction. Nature 2000, 407, 530–535. [Google Scholar] [CrossRef]
  85. Wehrli, M.; Dougan, S.T.; Caldwell, K.; O’Keefe, L.; Schwartz, S.; Vaizel-Ohayon, D.; Schejter, E.; Tomlinson, A.; DiNardo, S. Arrow Encodes an LDL-Receptor-Related Protein Essential for Wingless Signalling. Nature 2000, 407, 527–530. [Google Scholar] [CrossRef]
  86. Brunner, E.; Peter, O.; Schweizer, L.; Basler, K. Pangolinencodes a Lef-1 Homologue That Acts Downstream of Armadillo to Transduce the Wingless Signal in Drosophila. Nature 1997, 385, 829–833. [Google Scholar] [CrossRef]
  87. Zhang, X.; Gaspard, J.P.; Chung, D.C. Regulation of Vascular Endothelial Growth Factor by the Wnt and K-Ras Pathways in Colonic Neoplasia. Cancer Res. 2001, 61, 6050–6054. [Google Scholar] [PubMed]
  88. Inoki, K.; Ouyang, H.; Zhu, T.; Lindvall, C.; Wang, Y.; Zhang, X.; Yang, Q.; Bennett, C.; Harada, Y.; Stankunas, K.; et al. TSC2 Integrates Wnt and Energy Signals via a Coordinated Phosphorylation by AMPK and GSK3 to Regulate Cell Growth. Cell 2006, 126, 955–968. [Google Scholar] [CrossRef] [Green Version]
  89. Hey, P.J.; Twells, R.C.J.; Phillips, M.S.; Nakagawa, Y.; Brown, S.D.; Kawaguchi, Y.; Cox, R.; Xie, G.; Dugan, V.; Hammond, H.; et al. Cloning of a Novel Member of the Low-Density Lipoprotein Receptor Family1P.J. Hey and R.C.J. Twells Contributed Equally to This Work.1. Gene 1998, 216, 103–111. [Google Scholar] [CrossRef]
  90. Moon, R.T.; Kohn, A.D.; Ferrari, G.V.D.; Kaykas, A. WNT and β-Catenin Signalling: Diseases and Therapies. Nat. Rev. Genet. 2004, 5, 691–701. [Google Scholar] [CrossRef]
  91. Wodarz, A.; Nusse, R. Mechanisms of Wnt Signaling in Development. Annu. Rev. Cell Dev. Biol. 1998, 14, 59–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Lamb, R.; Ablett, M.P.; Spence, K.; Landberg, G.; Sims, A.H.; Clarke, R.B. Wnt Pathway Activity in Breast Cancer Sub-Types and Stem-like Cells. PLoS ONE 2013, 8, e67811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhan, T.; Ambrosi, G.; Wandmacher, A.M.; Rauscher, B.; Betge, J.; Rindtorff, N.; Häussler, R.S.; Hinsenkamp, I.; Bamberg, L.; Hessling, B.; et al. MEK Inhibitors Activate Wnt Signalling and Induce Stem Cell Plasticity in Colorectal Cancer. Nat. Commun 2019, 10, 2197. [Google Scholar] [CrossRef] [PubMed]
  94. Anastas, J.N.; Moon, R.T. WNT Signalling Pathways as Therapeutic Targets in Cancer. Nat. Rev. Cancer 2013, 13, 11–26. [Google Scholar] [CrossRef]
  95. Rabbani, S.A.; Arakelian, A.; Farookhi, R. LRP5 Knockdown: Effect on Prostate Cancer Invasion Growth and Skeletal Metastasis in Vitro and in Vivo. Cancer Med. 2013, 2, 625–635. [Google Scholar] [CrossRef]
  96. Hoang, B.H.; Kubo, T.; Healey, J.H.; Sowers, R.; Mazza, B.; Yang, R.; Huvos, A.G.; Meyers, P.A.; Gorlick, R. Expression of LDL Receptor-Related Protein 5 (LRP5) as a Novel Marker for Disease Progression in High-Grade Osteosarcoma. Int. J. Cancer 2004, 109, 106–111. [Google Scholar] [CrossRef]
  97. Liu, C.-C.; Prior, J.; Piwnica-Worms, D.; Bu, G. LRP6 Overexpression Defines a Class of Breast Cancer Subtype and Is a Target for Therapy. Proc. Natl. Acad. Sci. USA 2010, 107, 5136–5141. [Google Scholar] [CrossRef] [Green Version]
  98. Raisch, J.; Côté-Biron, A.; Rivard, N. A Role for the WNT Co-Receptor LRP6 in Pathogenesis and Therapy of Epithelial Cancers. Cancers 2019, 11, 1162. [Google Scholar] [CrossRef] [Green Version]
  99. Deng, D.; Zhang, Y.; Bao, W.; Kong, X. Low-Density Lipoprotein Receptor-Related Protein 6 (LRP6) Rs10845498 Polymorphism Is Associated with a Decreased Risk of Non-Small Cell Lung Cancer. Int. J. Med. Sci. 2014, 11, 685–690. [Google Scholar] [CrossRef] [Green Version]
  100. Tung, E.K.-K.; Wong, B.Y.-C.; Yau, T.-O.; Ng, I.O.-L. Upregulation of the Wnt Co-Receptor LRP6 Promotes Hepatocarcinogenesis and Enhances Cell Invasion. PLoS ONE 2012, 7, e36565. [Google Scholar] [CrossRef] [Green Version]
  101. Rismani, E.; Fazeli, M.S.; Mahmoodzadeh, H.; Movassagh, A.; Azami, S.; Karimipoor, M.; Teimoori-Toolabi, L. Pattern of LRP6 Gene Expression in Tumoral Tissues of Colorectal Cancer. Cancer Biomark. 2017, 19, 151–159. [Google Scholar] [CrossRef] [PubMed]
  102. Garg, B.; Giri, B.; Majumder, K.; Dudeja, V.; Banerjee, S.; Saluja, A. Modulation of Post-Translational Modifications in β-Catenin and LRP6 Inhibits Wnt Signaling Pathway in Pancreatic Cancer. Cancer Lett. 2017, 388, 64–72. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, J.; Li, Y.; Liu, Q.; Lu, W.; Bu, G. Wnt Signaling Activation and Mammary Gland Hyperplasia in MMTV–LRP6 Transgenic Mice: Implication for Breast Cancer Tumorigenesis. Oncogene 2010, 29, 539–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Liu, C.-C.; Tsai, C.-W.; Deak, F.; Rogers, J.; Penuliar, M.; Sung, Y.M.; Maher, J.N.; Fu, Y.; Li, X.; Xu, H.; et al. Deficiency in LRP6-Mediated Wnt Signaling Contributes to Synaptic Abnormalities and Amyloid Pathology in Alzheimer’s Disease. Neuron 2014, 84, 63–77. [Google Scholar] [CrossRef] [Green Version]
  105. Liu, C.X.; Li, Y.; Obermoeller-McCormick, L.M.; Schwartz, A.L.; Bu, G. The Putative Tumor Suppressor LRP1B, a Novel Member of the Low Density Lipoprotein (LDL) Receptor Family, Exhibits Both Overlapping and Distinct Properties with the LDL Receptor-Related Protein. J. Biol. Chem. 2001, 276, 28889–28896. [Google Scholar] [CrossRef] [Green Version]
  106. Li, Y.; Knisely, J.M.; Lu, W.; McCormick, L.M.; Wang, J.; Henkin, J.; Schwartz, A.L.; Bu, G. Low Density Lipoprotein (LDL) Receptor-Related Protein 1B Impairs Urokinase Receptor Regeneration on the Cell Surface and Inhibits Cell Migration. J. Biol. Chem. 2002, 277, 42366–42371. [Google Scholar] [CrossRef] [Green Version]
  107. Lu, Y.-J.; Wu, C.-S.; Li, H.-P.; Liu, H.-P.; Lu, C.-Y.; Leu, Y.-W.; Wang, C.-S.; Chen, L.-C.; Lin, K.-H.; Chang, Y.-S. Aberrant Methylation Impairs Low Density Lipoprotein Receptor-Related Protein 1B Tumor Suppressor Function in Gastric Cancer. Genes Chromosomes Cancer 2010, 49, 412–424. [Google Scholar] [CrossRef]
  108. Prazeres, H.; Torres, J.; Rodrigues, F.; Pinto, M.; Pastoriza, M.C.; Gomes, D.; Cameselle-Teijeiro, J.; Vidal, A.; Martins, T.C.; Sobrinho-Simões, M.; et al. Chromosomal, Epigenetic and MicroRNA-Mediated Inactivation of LRP1B, a Modulator of the Extracellular Environment of Thyroid Cancer Cells. Oncogene 2011, 30, 1302–1317. [Google Scholar] [CrossRef] [Green Version]
  109. Wang, Z.; Sun, P.; Gao, C.; Chen, J.; Li, J.; Chen, Z.; Xu, M.; Shao, J.; Zhang, Y.; Xie, J. Down-Regulation of LRP1B in Colon Cancer Promoted the Growth and Migration of Cancer Cells. Exp. Cell Res. 2017, 357, 1–8. [Google Scholar] [CrossRef]
  110. Langbein, S.; Szakacs, O.; Wilhelm, M.; Sukosd, F.; Weber, S.; Jauch, A.; Beltran, A.L.; Alken, P.; Kälble, T.; Kovacs, G. Alteration of the LRP1B Gene Region Is Associated with High Grade of Urothelial Cancer. Lab. Investig. 2002, 82, 639–643. [Google Scholar] [CrossRef] [Green Version]
  111. Sonoda, I.; Imoto, I.; Inoue, J.; Shibata, T.; Shimada, Y.; Chin, K.; Imamura, M.; Amagasa, T.; Gray, J.W.; Hirohashi, S.; et al. Frequent Silencing of Low Density Lipoprotein Receptor-Related Protein 1B (LRP1B) Expression by Genetic and Epigenetic Mechanisms in Esophageal Squamous Cell Carcinoma. Cancer Res. 2004, 64, 3741–3747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Rahmatpanah, F.B.; Carstens, S.; Guo, J.; Sjahputera, O.; Taylor, K.H.; Duff, D.; Shi, H.; Davis, J.W.; Hooshmand, S.I.; Chitma-Matsiga, R.; et al. Differential DNA Methylation Patterns of Small B-Cell Lymphoma Subclasses with Different Clinical Behavior. Leukemia 2006, 20, 1855–1862. [Google Scholar] [CrossRef] [PubMed]
  113. Taylor, K.H.; Pena-Hernandez, K.E.; Davis, J.W.; Arthur, G.L.; Duff, D.J.; Shi, H.; Rahmatpanah, F.B.; Sjahputera, O.; Caldwell, C.W. Large-Scale CpG Methylation Analysis Identifies Novel Candidate Genes and Reveals Methylation Hotspots in Acute Lymphoblastic Leukemia. Cancer Res. 2007, 67, 2617–2625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ding, L.; Getz, G.; Wheeler, D.A.; Mardis, E.R.; McLellan, M.D.; Cibulskis, K.; Sougnez, C.; Greulich, H.; Muzny, D.M.; Morgan, M.B.; et al. Somatic Mutations Affect Key Pathways in Lung Adenocarcinoma. Nature 2008, 455, 1069–1075. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, C.X.; Musco, S.; Lisitsina, N.M.; Yaklichkin, S.Y.; Lisitsyn, N.A. Genomic Organization of a New Candidate Tumor Suppressor Gene, LRP1B. Genomics 2000, 69, 271–274. [Google Scholar] [CrossRef] [PubMed]
  116. Marschang, P.; Brich, J.; Weeber, E.J.; Sweatt, J.D.; Shelton, J.M.; Richardson, J.A.; Hammer, R.E.; Herz, J. Normal Development and Fertility of Knockout Mice Lacking the Tumor Suppressor Gene LRP1b Suggest Functional Compensation by LRP1. Mol. Cell Biol. 2004, 24, 3782–3793. [Google Scholar] [CrossRef] [Green Version]
  117. Marzolo, M.P.; von Bernhardi, R.; Bu, G.; Inestrosa, N.C. Expression of Alpha(2)-Macroglobulin Receptor/Low Density Lipoprotein Receptor-Related Protein (LRP) in Rat Microglial Cells. J. Neurosci. Res. 2000, 60, 401–411. [Google Scholar] [CrossRef]
  118. Cam, J.A.; Zerbinatti, C.V.; Knisely, J.M.; Hecimovic, S.; Li, Y.; Bu, G. The Low Density Lipoprotein Receptor-Related Protein 1B Retains Beta-Amyloid Precursor Protein at the Cell Surface and Reduces Amyloid-Beta Peptide Production. J. Biol. Chem. 2004, 279, 29639–29646. [Google Scholar] [CrossRef] [Green Version]
  119. Arnaud, L.; Ballif, B.A.; Förster, E.; Cooper, J.A. Fyn Tyrosine Kinase Is a Critical Regulator of Disabled-1 during Brain Development. Curr. Biol. 2003, 13, 9–17. [Google Scholar] [CrossRef] [Green Version]
  120. Kim, D.-H.; Iijima, H.; Goto, K.; Sakai, J.; Ishii, H.; Kim, H.-J.; Suzuki, H.; Kondo, H.; Saeki, S.; Yamamoto, T. Human Apolipoprotein E Receptor 2: A Novel Lipoprotein Receptor of the Low Density Lipoprotein Receptor Family Predominantly Expressed In Brain. J. Biol. Chem. 1996, 271, 8373–8380. [Google Scholar] [CrossRef] [Green Version]
  121. Strøm, T.B.; Tveten, K.; Holla, Ø.L.; Cameron, J.; Berge, K.E.; Leren, T.P. Characterization of Residues in the Cytoplasmic Domain of the LDL Receptor Required for Exit from the Endoplasmic Reticulum. Biochem. Biophys. Res. Commun. 2011, 415, 642–645. [Google Scholar] [CrossRef] [PubMed]
  122. Uhlik, M.T.; Temple, B.; Bencharit, S.; Kimple, A.J.; Siderovski, D.P.; Johnson, G.L. Structural and Evolutionary Division of Phosphotyrosine Binding (PTB) Domains. J. Mol. Biol 2005, 345, 1–20. [Google Scholar] [CrossRef] [PubMed]
  123. Balmaceda, V.; Cuchillo-Ibáñez, I.; Pujadas, L.; García-Ayllón, M.-S.; Saura, C.A.; Nimpf, J.; Soriano, E.; Sáez-Valero, J. ApoER2 Processing by Presenilin-1 Modulates Reelin Expression. FASEB J. 2014, 28, 1543–1554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Telese, F.; Ma, Q.; Perez, P.M.; Notani, D.; Oh, S.; Li, W.; Comoletti, D.; Ohgi, K.A.; Taylor, H.; Rosenfeld, M.G. LRP8-Reelin-Regulated Neuronal Enhancer Signature Underlying Learning and Memory Formation. Neuron 2015, 86, 696–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Sun, X.-M.; Soutar, A.K. Expression in Vitro of Alternatively Spliced Variants of the Messenger RNA for Human Apolipoprotein E Receptor-2 Identified in Human Tissues by Ribonuclease Protection Assays. Eur. J. Biochem. 1999, 262, 230–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Clatworthy, A.E.; Stockinger, W.; Christie, R.H.; Schneider, W.J.; Nimpf, J.; Hyman, B.T.; Rebeck, G.W. Expression and Alternate Splicing of Apolipoprotein E Receptor 2 in Brain. Neuroscience 1999, 90, 903–911. [Google Scholar] [CrossRef]
  127. Gallo, C.M.; Labadorf, A.T.; Ho, A.; Beffert, U. Single Molecule, Long-Read Apoer2 Sequencing Identifies Conserved and Species-Specific Splicing Patterns. Genomics 2022, 114, 110318. [Google Scholar] [CrossRef]
  128. Hinrich, A.J.; Jodelka, F.M.; Chang, J.L.; Brutman, D.; Bruno, A.M.; Briggs, C.A.; James, B.D.; Stutzmann, G.E.; Bennett, D.A.; Miller, S.A.; et al. Therapeutic Correction of ApoER2 Splicing in Alzheimer’s Disease Mice Using Antisense Oligonucleotides. EMBO Mol. Med. 2016, 8, 328–345. [Google Scholar] [CrossRef] [Green Version]
  129. Brandes, C.; Kahr, L.; Stockinger, W.; Hiesberger, T.; Schneider, W.J.; Nimpf, J. Alternative Splicing in the Ligand Binding Domain of Mouse ApoE Receptor-2 Produces Receptor Variants Binding Reelin but Not Alpha 2-Macroglobulin. J. Biol. Chem. 2001, 276, 22160–22169. [Google Scholar] [CrossRef] [Green Version]
  130. Wasser, C.R.; Masiulis, I.; Durakoglugil, M.S.; Lane-Donovan, C.; Xian, X.; Beffert, U.; Agarwala, A.; Hammer, R.E.; Herz, J. Differential Splicing and Glycosylation of Apoer2 Alters Synaptic Plasticity and Fear Learning. Sci. Signal. 2014, 7, ra113. [Google Scholar] [CrossRef] [Green Version]
  131. Beffert, U.; Nematollah Farsian, F.; Masiulis, I.; Hammer, R.E.; Yoon, S.O.; Giehl, K.M.; Herz, J. ApoE Receptor 2 Controls Neuronal Survival in the Adult Brain. Curr. Biol. 2006, 16, 2446–2452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Stockinger, W.; Hengstschläger-Ottnad, E.; Novak, S.; Matus, A.; Hüttinger, M.; Bauer, J.; Lassmann, H.; Schneider, W.J.; Nimpf, J. The Low Density Lipoprotein Receptor Gene Family. Differential Expression of Two Alpha2-Macroglobulin Receptors in the Brain. J. Biol. Chem. 1998, 273, 32213–32221. [Google Scholar] [CrossRef] [Green Version]
  133. Brandes, C.; Novak, S.; Stockinger, W.; Herz, J.; Schneider, W.J.; Nimpf, J. Avian and Murine LR8B and Human Apolipoprotein E Receptor 2: Differentially Spliced Products from Corresponding Genes. Genomics 1997, 42, 185–191. [Google Scholar] [CrossRef] [PubMed]
  134. Myant, N.B. Reelin and Apolipoprotein E Receptor 2 in the Embryonic and Mature Brain: Effects of an Evolutionary Change in the ApoER2 Gene. Proc. Biol. Sci. 2010, 277, 345–351. [Google Scholar] [CrossRef] [Green Version]
  135. Omuro, K.C.; Gallo, C.M.; Scrandis, L.; Ho, A.; Beffert, U. Human APOER2 Isoforms Have Differential Cleavage Events and Synaptic Properties. J. Neurosci. 2022, 42, 4054–4068. [Google Scholar] [CrossRef]
  136. Reddy, S.S.; Connor, T.E.; Weeber, E.J.; Rebeck, W. Similarities and Differences in Structure, Expression, and Functions of VLDLR and ApoER2. Mol. Neurodegener. 2011, 6, 30. [Google Scholar] [CrossRef] [Green Version]
  137. Pasten, C.; Cerda, J.; Jausoro, I.; Court, F.A.; Cáceres, A.; Marzolo, M.-P. ApoER2 and Reelin Are Expressed in Regenerating Peripheral Nerve and Regulate Schwann Cell Migration by Activating the Rac1 GEF Protein, Tiam1. Mol. Cell Neurosci. 2015, 69, 1–11. [Google Scholar] [CrossRef] [PubMed]
  138. Waltmann, M.D.; Basford, J.E.; Konaniah, E.S.; Weintraub, N.L.; Hui, D.Y. Apolipoprotein E Receptor-2 Deficiency Enhances Macrophage Susceptibility to Lipid Accumulation and Cell Death to Augment Atherosclerotic Plaque Progression and Necrosis. Biochim. Biophys. Acta 2014, 1842, 1395–1405. [Google Scholar] [CrossRef] [Green Version]
  139. Ulrich, V.; Konaniah, E.S.; Herz, J.; Gerard, R.D.; Jung, E.; Yuhanna, I.S.; Ahmed, M.; Hui, D.Y.; Mineo, C.; Shaul, P.W. Genetic Variants of ApoE and ApoER2 Differentially Modulate Endothelial Function. Proc. Natl. Acad. Sci. USA 2014, 111, 13493–13498. [Google Scholar] [CrossRef] [Green Version]
  140. Huang, J.; Li, L.; Lian, J.; Schauer, S.; Vesely, P.W.; Kratky, D.; Hoefler, G.; Lehner, R. Tumor-Induced Hyperlipidemia Contributes to Tumor Growth. Cell Rep. 2016, 15, 336–348. [Google Scholar] [CrossRef] [Green Version]
  141. Zheng, S.; Wei, Y.; Jiang, Y.; Hao, Y. LRP8 Activates STAT3 to Induce PD-L1 Expression in Osteosarcoma. Tumori 2021, 107, 238–246. [Google Scholar] [CrossRef] [PubMed]
  142. Maire, V.; Mahmood, F.; Rigaill, G.; Ye, M.; Brisson, A.; Némati, F.; Gentien, D.; Tucker, G.C.; Roman-Roman, S.; Dubois, T. LRP8 Is Overexpressed in Estrogen-Negative Breast Cancers and a Potential Target for These Tumors. Cancer Med. 2019, 8, 325–336. [Google Scholar] [CrossRef] [PubMed]
  143. Dun, B.; Sharma, A.; Teng, Y.; Liu, H.; Purohit, S.; Xu, H.; Zeng, L.; She, J.-X. Mycophenolic Acid Inhibits Migration and Invasion of Gastric Cancer Cells via Multiple Molecular Pathways. PLoS ONE 2013, 8, e81702. [Google Scholar] [CrossRef] [PubMed]
  144. Lu, J.; Ma, Y.; Zhao, Z. MiR-142 Suppresses Progression of Gastric Carcinoma via Directly Targeting LRP8. Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101520. [Google Scholar] [CrossRef]
  145. Cai, J.; Chen, J.; Wu, T.; Cheng, Z.; Tian, Y.; Pu, C.; Shi, W.; Suo, X.; Wu, X.; Zhang, K. Genome-Scale CRISPR Activation Screening Identifies a Role of LRP8 in Sorafenib Resistance in Hepatocellular Carcinoma. Biochem. Biophys. Res. Commun. 2020, 526, 1170–1176. [Google Scholar] [CrossRef]
  146. Pencheva, N.; Tran, H.; Buss, C.; Huh, D.; Drobnjak, M.; Busam, K.; Tavazoie, S.F. Convergent Multi-MiRNA Targeting of ApoE Drives LRP1/LRP8-Dependent Melanoma Metastasis and Angiogenesis. Cell 2012, 151, 1068–1082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Qiu, H.; Shen, X.; Chen, B.; Chen, T.; Feng, G.; Chen, S.; Feng, D.; Xu, Q. MiR-30b-5p Inhibits Cancer Progression and Enhances Cisplatin Sensitivity in Lung Cancer through Targeting LRP8. Apoptosis 2021, 26, 261–276. [Google Scholar] [CrossRef]
  148. Fang, Z.; Zhong, M.; Zhou, L.; Le, Y.; Wang, H.; Fang, Z. Low-Density Lipoprotein Receptor-Related Protein 8 Facilitates the Proliferation and Invasion of Non-Small Cell Lung Cancer Cells by Regulating the Wnt/β-Catenin Signaling Pathway. Bioengineered 2022, 13, 6807–6818. [Google Scholar] [CrossRef]
  149. Arai, T.; Kojima, S.; Yamada, Y.; Sugawara, S.; Kato, M.; Yamazaki, K.; Naya, Y.; Ichikawa, T.; Seki, N. Pirin: A Potential Novel Therapeutic Target for Castration-resistant Prostate Cancer Regulated by MiR-455-5p. Mol. Oncol. 2019, 13, 322–337. [Google Scholar] [CrossRef] [Green Version]
  150. Du, S.; Wang, H.; Cai, J.; Ren, R.; Zhang, W.; Wei, W.; Shen, X. Apolipoprotein E2 Modulates Cell Cycle Function to Promote Proliferation in Pancreatic Cancer Cells via Regulation of the C-Myc–P21 Waf1 Signalling Pathway. Biochem. Cell Biol. 2020, 98, 191–202. [Google Scholar] [CrossRef]
  151. Zhang, J.; Zhang, X.; Zhang, L.; Zhou, F.; van Dinther, M.; Ten Dijke, P. LRP8 Mediates Wnt/β-Catenin Signaling and Controls Osteoblast Differentiation. J. Bone Miner. Res. 2012, 27, 2065–2074. [Google Scholar] [CrossRef]
  152. Poirier, S.; Mayer, G.; Benjannet, S.; Bergeron, E.; Marcinkiewicz, J.; Nassoury, N.; Mayer, H.; Nimpf, J.; Prat, A.; Seidah, N.G. The Proprotein Convertase PCSK9 Induces the Degradation of Low Density Lipoprotein Receptor (LDLR) and Its Closest Family Members VLDLR and ApoER2. J. Biol. Chem. 2008, 283, 2363–2372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Huang, X.; Zhang, W.; Zhang, Z.; Shi, D.; Wu, F.; Zhong, B.; Shao, Z. Prognostic Value of Programmed Cell Death 1 Ligand-1 (PD-L1) or PD-1 Expression in Patients with Osteosarcoma: A Meta-Analysis. J. Cancer 2018, 9, 2525–2531. [Google Scholar] [CrossRef] [PubMed]
  154. Seidah, N.G. The PCSK9 Revolution and the Potential of PCSK9-Based Therapies to Reduce LDL-Cholesterol. Glob. Cardiol. Sci. Pract. 2017, 2017, e201702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Ruvolo, P.P. The Broken “Off” Switch in Cancer Signaling: PP2A as a Regulator of Tumorigenesis, Drug Resistance, and Immune Surveillance. BBA Clin. 2016, 6, 87–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Komaravolu, R.K.; Waltmann, M.D.; Konaniah, E.; Jaeschke, A.; Hui, D.Y. ApoER2 (Apolipoprotein E Receptor-2) Deficiency Accelerates Smooth Muscle Cell Senescence via Cytokinesis Impairment and Promotes Fibrotic Neointima After Vascular Injury. ATVB 2019, 39, 2132–2144. [Google Scholar] [CrossRef] [PubMed]
  157. He, X.; Cooley, K.; Chung, C.H.Y.; Dashti, N.; Tang, J. Apolipoprotein Receptor 2 and X11 Alpha/Beta Mediate Apolipoprotein E-Induced Endocytosis of Amyloid-Beta Precursor Protein and Beta-Secretase, Leading to Amyloid-Beta Production. J. Neurosci. 2007, 27, 4052–4060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Hoe, H.-S.; Wessner, D.; Beffert, U.; Becker, A.G.; Matsuoka, Y.; Rebeck, G.W. F-Spondin Interaction with the Apolipoprotein E Receptor ApoEr2 Affects Processing of Amyloid Precursor Protein. Mol. Cell Biol. 2005, 25, 9259–9268. [Google Scholar] [CrossRef] [Green Version]
  159. Fuentealba, R.A.; Barría, M.I.; Lee, J.; Cam, J.; Araya, C.; Escudero, C.A.; Inestrosa, N.C.; Bronfman, F.C.; Bu, G.; Marzolo, M.-P. ApoER2 Expression Increases Abeta Production While Decreasing Amyloid Precursor Protein (APP) Endocytosis: Possible Role in the Partitioning of APP into Lipid Rafts and in the Regulation of Gamma-Secretase Activity. Mol. Neurodegener. 2007, 2, 14. [Google Scholar] [CrossRef] [Green Version]
  160. Bock, H.H.; Herz, J. Reelin Activates SRC Family Tyrosine Kinases in Neurons. Curr. Biol. 2003, 13, 18–26. [Google Scholar] [CrossRef] [Green Version]
  161. Beffert, U.; Morfini, G.; Bock, H.H.; Reyna, H.; Brady, S.T.; Herz, J. Reelin-Mediated Signaling Locally Regulates Protein Kinase B/Akt and Glycogen Synthase Kinase 3beta. J. Biol. Chem. 2002, 277, 49958–49964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Meyer, D.; Liu, A.; Margolis, B. Interaction of C-Jun Amino-Terminal Kinase Interacting Protein-1 with P190 RhoGEF and Its Localization in Differentiated Neurons. J. Biol. Chem. 1999, 274, 35113–35118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Chen, Y. Reelin Modulates NMDA Receptor Activity in Cortical Neurons. J. Neurosci. 2005, 25, 8209–8216. [Google Scholar] [CrossRef] [PubMed]
  164. Roche, K.W.; Standley, S.; McCallum, J.; Dune Ly, C.; Ehlers, M.D.; Wenthold, R.J. Molecular Determinants of NMDA Receptor Internalization. Nat. Neurosci. 2001, 4, 794–802. [Google Scholar] [CrossRef] [PubMed]
  165. Srivastava, R.A. Regulation of the Apolipoprotein E by Dietary Lipids Occurs by Transcriptional and Post-Transcriptional Mechanisms. Mol. Cell. Biochem. 1996, 155, 153–162. [Google Scholar] [CrossRef]
  166. Rall, S.C.; Weisgraber, K.H.; Mahley, R.W. Human Apolipoprotein E. The Complete Amino Acid Sequence. J. Biol. Chem. 1982, 257, 4171–4178. [Google Scholar] [CrossRef]
  167. Abubakar, F.; Iqbal, M.P. Apolipoprotein E (Apo E) Gene Polymorphism and Coronary Heart Disease in Asian Populations. Pak. J. Pharm. Sci. 2015, 28, 1439–1444. [Google Scholar]
  168. Fan, Y.-Y.; Cai, Q.-L.; Gao, Z.-Y.; Lin, X.; Huang, Q.; Tang, W.; Liu, J.-H. APOE Ε4 Allele Elevates the Expressions of Inflammatory Factors and Promotes Alzheimer’s Disease Progression: A Comparative Study Based on Han and She Populations in the Wenzhou Area. Brain Res. Bull. 2017, 132, 39–43. [Google Scholar] [CrossRef]
  169. Corder, E.H.; Saunders, A.M.; Strittmatter, W.J.; Schmechel, D.E.; Gaskell, P.C.; Small, G.W.; Roses, A.D.; Haines, J.L.; Pericak-Vance, M.A. Gene Dose of Apolipoprotein E Type 4 Allele and the Risk of Alzheimer’s Disease in Late Onset Families. Science 1993, 261, 921–923. [Google Scholar] [CrossRef]
  170. Holtzman, D.M.; Herz, J.; Bu, G. Apolipoprotein E and Apolipoprotein E Receptors: Normal Biology and Roles in Alzheimer Disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Gordon, I.; Grauer, E.; Genis, I.; Sehayek, E.; Michaelson, D.M. Memory Deficits and Cholinergic Impairments in Apolipoprotein E-Deficient Mice. Neurosci. Lett. 1995, 199, 1–4. [Google Scholar] [CrossRef]
  172. Masliah, E.; Mallory, M.; Ge, N.; Alford, M.; Veinbergs, I.; Roses, A.D. Neurodegeneration in the Central Nervous System of ApoE-Deficient Mice. Exp. Neurol. 1995, 136, 107–122. [Google Scholar] [CrossRef]
  173. Davis, A.A.; Inman, C.E.; Wargel, Z.M.; Dube, U.; Freeberg, B.M.; Galluppi, A.; Haines, J.N.; Dhavale, D.D.; Miller, R.; Choudhury, F.A.; et al. APOE Genotype Regulates Pathology and Disease Progression in Synucleinopathy. Sci. Transl. Med. 2020, 12, eaay3069. [Google Scholar] [CrossRef] [PubMed]
  174. Tsuang, D.; Leverenz, J.B.; Lopez, O.L.; Hamilton, R.L.; Bennett, D.A.; Schneider, J.A.; Buchman, A.S.; Larson, E.B.; Crane, P.K.; Kaye, J.A.; et al. APOE Ε4 Increases Risk for Dementia in Pure Synucleinopathies. JAMA Neurol. 2013, 70, 223–228. [Google Scholar] [CrossRef] [Green Version]
  175. Aleshkov, S.; Abraham, C.R.; Zannis, V.I. Interaction of Nascent ApoE2, ApoE3, and ApoE4 Isoforms Expressed in Mammalian Cells with Amyloid Peptide Beta (1-40). Relevance to Alzheimer’s Disease. Biochemistry 1997, 36, 10571–10580. [Google Scholar] [CrossRef]
  176. Petrlova, J.; Hong, H.-S.; Bricarello, D.A.; Harishchandra, G.; Lorigan, G.A.; Jin, L.-W.; Voss, J.C. A Differential Association of Apolipoprotein E Isoforms with the Amyloid-β Oligomer in Solution. Proteins 2011, 79, 402–416. [Google Scholar] [CrossRef] [Green Version]
  177. Bales, K.R.; Verina, T.; Dodel, R.C.; Du, Y.; Altstiel, L.; Bender, M.; Hyslop, P.; Johnstone, E.M.; Little, S.P.; Cummins, D.J.; et al. Lack of Apolipoprotein E Dramatically Reduces Amyloid Beta-Peptide Deposition. Nat. Genet. 1997, 17, 263–264. [Google Scholar] [CrossRef] [PubMed]
  178. Irizarry, M.C.; Rebeck, G.W.; Cheung, B.; Bales, K.; Paul, S.M.; Holzman, D.; Hyman, B.T. Modulation of A Beta Deposition in APP Transgenic Mice by an Apolipoprotein E Null Background. Ann. N. Y. Acad. Sci. 2000, 920, 171–178. [Google Scholar] [CrossRef]
  179. May, P.; Reddy, Y.K.; Herz, J. Proteolytic Processing of Low Density Lipoprotein Receptor-Related Protein Mediates Regulated Release of Its Intracellular Domain. J. Biol. Chem. 2002, 277, 18736–18743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. 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] [CrossRef] [Green Version]
  181. Nizzari, M.; Thellung, S.; Corsaro, A.; Villa, V.; Pagano, A.; Porcile, C.; Russo, C.; Florio, T. Neurodegeneration in Alzheimer Disease: Role of Amyloid Precursor Protein and Presenilin 1 Intracellular Signaling. J. Toxicol. 2012, 2012, 187297. [Google Scholar] [CrossRef] [PubMed]
  182. Nizzari, M.; Barbieri, F.; Gentile, M.T.; Passarella, D.; Caorsi, C.; Diaspro, A.; Taglialatela, M.; Pagano, A.; Colucci-D’Amato, L.; Florio, T.; et al. Amyloid-β Protein Precursor Regulates Phosphorylation and Cellular Compartmentalization of Microtubule Associated Protein Tau. J. Alzheimers Dis. 2012, 29, 211–227. [Google Scholar] [CrossRef] [PubMed]
  183. D’Arcangelo, G.; Homayouni, R.; Keshvara, L.; Rice, D.S.; Sheldon, M.; Curran, T. Reelin Is a Ligand for Lipoprotein Receptors. Neuron 1999, 24, 471–479. [Google Scholar] [CrossRef] [Green Version]
  184. Beffert, U.; Weeber, E.J.; Morfini, G.; Ko, J.; Brady, S.T.; Tsai, L.-H.; Sweatt, J.D.; Herz, J. Reelin and Cyclin-Dependent Kinase 5-Dependent Signals Cooperate in Regulating Neuronal Migration and Synaptic Transmission. J. Neurosci. 2004, 24, 1897–1906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Hoe, H.-S.; Lee, K.J.; Carney, R.S.E.; Lee, J.; Markova, A.; Lee, J.-Y.; Howell, B.W.; Hyman, B.T.; Pak, D.T.S.; Bu, G.; et al. Interaction of Reelin with Amyloid Precursor Protein Promotes Neurite Outgrowth. J. Neurosci. 2009, 29, 7459–7473. [Google Scholar] [CrossRef] [PubMed]
  186. Niu, S.; Yabut, O.; D’Arcangelo, G. The Reelin Signaling Pathway Promotes Dendritic Spine Development in Hippocampal Neurons. J. Neurosci. 2008, 28, 10339–10348. [Google Scholar] [CrossRef]
  187. Beffert, U.; Weeber, E.J.; Durudas, A.; Qiu, S.; Masiulis, I.; Sweatt, J.D.; Li, W.-P.; Adelmann, G.; Frotscher, M.; Hammer, R.E.; et al. Modulation of Synaptic Plasticity and Memory by Reelin Involves Differential Splicing of the Lipoprotein Receptor Apoer2. Neuron 2005, 47, 567–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Herz, J.; Chen, Y. Reelin, Lipoprotein Receptors and Synaptic Plasticity. Nat. Rev. Neurosci. 2006, 7, 850–859. [Google Scholar] [CrossRef] [PubMed]
  189. Chen, K.; Ochalski, P.G.; Tran, T.S.; Sahir, N.; Schubert, M.; Pramatarova, A.; Howell, B.W. Interaction between Dab1 and CrkII Is Promoted by Reelin Signaling. J. Cell. Sci. 2004, 117, 4527–4536. [Google Scholar] [CrossRef] [Green Version]
  190. Parent, A.T.; Thinakaran, G. Modeling Presenilin-Dependent Familial Alzheimer’s Disease: Emphasis on Presenilin Substrate-Mediated Signaling and Synaptic Function. Int. J. Alzheimers Dis. 2010, 2010, 825918. [Google Scholar] [CrossRef] [Green Version]
  191. Raihan, O.; Brishti, A.; Li, Q.; Zhang, Q.; Li, D.; Li, X.; Zhang, Q.; Xie, Z.; Li, J.; Zhang, J.; et al. SFRS11 Loss Leads to Aging-Associated Cognitive Decline by Modulating LRP8 and ApoE. Cell. Rep. 2019, 28, 78–90.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Wang, W.; Moerman-Herzog, A.M.; Slaton, A.; Barger, S.W. Presenilin 1 Mutations Influence Processing and Trafficking of the ApoE Receptor ApoER2. Neurobiol. Aging 2017, 49, 145–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Blake, S.M.; Strasser, V.; Andrade, N.; Duit, S.; Hofbauer, R.; Schneider, W.J.; Nimpf, J. Thrombospondin-1 Binds to ApoER2 and VLDL Receptor and Functions in Postnatal Neuronal Migration. EMBO J. 2008, 27, 3069–3080. [Google Scholar] [CrossRef]
  194. Leeb, C.; Eresheim, C.; Nimpf, J. Clusterin Is a Ligand for Apolipoprotein E Receptor 2 (ApoER2) and Very Low Density Lipoprotein Receptor (VLDLR) and Signals via the Reelin-Signaling Pathway. J. Biol. Chem. 2014, 289, 4161–4172. [Google Scholar] [CrossRef] [Green Version]
  195. Zhang, Z.-H.; Song, G.-L. Roles of Selenoproteins in Brain Function and the Potential Mechanism of Selenium in Alzheimer’s Disease. Front. Neurosci. 2021, 15, 646518. [Google Scholar] [CrossRef] [PubMed]
  196. Gao, S.; Jin, Y.; Hall, K.S.; Liang, C.; Unverzagt, F.W.; Ji, R.; Murrell, J.R.; Cao, J.; Shen, J.; Ma, F.; et al. Selenium Level and Cognitive Function in Rural Elderly Chinese. Am. J. Epidemiol. 2007, 165, 955–965. [Google Scholar] [CrossRef] [Green Version]
  197. Berry, M.J.; Banu, L.; Harney, J.W.; Larsen, P.R. Functional Characterization of the Eukaryotic SECIS Elements Which Direct Selenocysteine Insertion at UGA Codons. EMBO J. 1993, 12, 3315–3322. [Google Scholar] [CrossRef]
  198. Chen, J.; Berry, M.J. Selenium and Selenoproteins in the Brain and Brain Diseases. J. Neurochem. 2003, 86, 1–12. [Google Scholar] [CrossRef]
  199. Burk, R.F.; Hill, K.E.; Olson, G.E.; Weeber, E.J.; Motley, A.K.; Winfrey, V.P.; Austin, L.M. Deletion of Apolipoprotein E Receptor-2 in Mice Lowers Brain Selenium and Causes Severe Neurological Dysfunction and Death When a Low-Selenium Diet Is Fed. J. Neurosci. 2007, 27, 6207–6211. [Google Scholar] [CrossRef]
  200. Olson, G.E.; Winfrey, V.P.; NagDas, S.K.; Hill, K.E.; Burk, R.F. Apolipoprotein E Receptor-2 (ApoER2) Mediates Selenium Uptake from Selenoprotein P by the Mouse Testis. J. Biol. Chem. 2007, 282, 12290–12297. [Google Scholar] [CrossRef] [Green Version]
  201. Medoro, A.; Bartollino, S.; Mignogna, D.; Passarella, D.; Porcile, C.; Pagano, A.; Florio, T.; Nizzari, M.; Guerra, G.; Di Marco, R.; et al. Complexity and Selectivity of γ-Secretase Cleavage on Multiple Substrates: Consequences in Alzheimer’s Disease and Cancer. J. Alzheimers Dis. 2018, 61, 1–15. [Google Scholar] [CrossRef] [PubMed]
  202. Medoro, A.; Bartollino, S.; Mignogna, D.; Marziliano, N.; Porcile, C.; Nizzari, M.; Florio, T.; Pagano, A.; Raimo, G.; Intrieri, M.; et al. Proteases Upregulation in Sporadic Alzheimer’s Disease Brain. J. Alzheimers Dis. 2019, 68, 931–938. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The domain structure of the LDL family members: differences and similarities. LDLR, VLDLR, and LRP8 are made of seven LA repeats in the ligand-binding domain, three EGF-like domains, and one YWTD β-propeller. LRP1 and LRP2 have the largest extracellular domains, each with eight YWTD β-propellers spaced by EGF and LA repeats. LRP4, LRP5 and LRP6 carry high sequence homology to a region within LRP1 with YWTD β-repeats going three through six. SorLA has two additional domains: the FNIII domain and the VPS10 domain. Regarding the NPxY motif, VLDLR, LRP8 and LR11/SorLA-1A have a single copy, whereas LRP1 and LRP1B contain two copies and LRP2/Megalin three copies. No NPXY is present in LRP5 and LRP6. Domains are not drawn to scale.
Figure 1. The domain structure of the LDL family members: differences and similarities. LDLR, VLDLR, and LRP8 are made of seven LA repeats in the ligand-binding domain, three EGF-like domains, and one YWTD β-propeller. LRP1 and LRP2 have the largest extracellular domains, each with eight YWTD β-propellers spaced by EGF and LA repeats. LRP4, LRP5 and LRP6 carry high sequence homology to a region within LRP1 with YWTD β-repeats going three through six. SorLA has two additional domains: the FNIII domain and the VPS10 domain. Regarding the NPxY motif, VLDLR, LRP8 and LR11/SorLA-1A have a single copy, whereas LRP1 and LRP1B contain two copies and LRP2/Megalin three copies. No NPXY is present in LRP5 and LRP6. Domains are not drawn to scale.
Ijms 23 08921 g001
Table 2. LRP8 in cancer-related molecular pathways. The table summarizes LRP8 main findings in cancer, with the corresponding roles, molecular mechanisms and in vitro and in vivo models used (see references).
Table 2. LRP8 in cancer-related molecular pathways. The table summarizes LRP8 main findings in cancer, with the corresponding roles, molecular mechanisms and in vitro and in vivo models used (see references).
Cancer TypesLRP8 Cancer
Roles
Main Findings &
Molecular Mechanism
In Vitro E in Vivo
Models
Refs.
OsteosarcomaCell proliferation
and
anti-apoptotic effect
LRP8 is overexpressed
in osteosarcoma tissues.
LRP8 enhances PD-L1 expression
via STAT3, evading the host
immune system.
Cell lines:
MG63
and U2OS
[141]
TNBC
TNBC
Cell proliferation,
anti-apoptotic effect and colony formation
LRP8 is overexpressed
in TNBC patients.
LRP8 depletion induces arrest
of the cell cycle and
apoptosis.
LRP8 knockdown impairs
colony formation.
Cell lines: BT-474, T47D, MCF7, ZR-75-1, SKBR3, HCC1569, HCC1954, BT-20, HCC1143, HCC38, HCC70, MDA-MB-468 and MDA-MB-45
In vivo model: xenograft mice model (MDA-MB-468)
[142]
Tumorigenesis
and
chemoresistance
LRP8 silencing suppresses BCSCs and tumorigenesis in TNBC via Wnt signaling inhibition.
LRP8 KO shifts TNBC cells to a more differentiated phenotype, sensitizing them to chemotherapy.
Cell lines:
HCC1937
and SUM149
In vivo model: xenograft NOD/SCID mice (SUM149)
[9]
Gastric
cancer
Cell migration Mycophenolic acid downregulates LRP8, reducing cell migration.Cell lines:
AGS and Hs746T
[143]
Cancer progression MiR-142 suppresses
progression of gastric carcinoma
via directly targeting LRP8.
Cell lines:
AGS, MKN-45, MKN-28, SGC-7901 and BGC-823
[144]
Hepatocellular
carcinoma
Pharmacoresistance LRP8-dependent activation of
β-catenin pathway suppresses
Sorafenib induced apoptosis.
Cell lines:
Huh7
and MHCC-97H
[145]
MelanomaSuppression of cell invasion and endothelial recruitment miR-1908, miR-199a-5p,
and miR-199a-3p
limit ApoE secretion suppressing
LRP8 endothelial engagement
Cell lines: TWM-266-4, A375, SK-Mel-28, HT-144, A2058, MeWo, SK-Mel-2, SK-Mel-28, A375, WM-266-4, HT-144, and A2058
In vivo model: xenograft NOD scid mice (MeWo)
[146]
Lung
cancer
Cancer progression
and
cisplatin resistance
miR-30b-5p inhibits
lung cancer cell viability,
migration and invasion and
enhances cell sensitivity
to DDP via targeting LRP8.
Cell lines:
A549, A549/DDP,
NCI-H1299, NCIH446
and H1650
In vivo model: xenograft BALB/c nude mice
[147]
Cell proliferation, migration, invasion, EMT, tumor growth
(NSCLC)
LRP8 is markedly overexpressed in NSCLC patients with poor clinicopathological characteristics and prognosis.
LRP8 KO elicits tumor-suppressive functions by suppressing the Wnt/β-catenin pathway.
Cell lines:
95-D, H1299, H460,
HCC-827, A549,
PC-9, and H1975
[148]
Prostate
cancer
Cancer progression miR-455-5p inhibits cancer cell
migration and invasive abilities
through LRP8 downregulation.
Cell lines:
PC3, DU145,
and C4-2
[149]
Pancreatic
cancer
Cell proliferation ApoE2-LRP8 induces phosphorylation
of ERK1/2 to activate c-Myc, promoting
cyclin D1, cdc2 and cyclin B1
expression and reducing p21Waf1 activity.
Cell lines: MIA PaCa-2, Capan-2, PANC-1, Bxpc-3 [150]
TNBC, Triple-negative breast cancer; BCSCs, Breast cancer stem cells; EMT, Epithelial-to-mesenchymal transition; NSCLC, Non-small cell lung cancer.
Table 3. LRP8 in CNS- and neurodegeneration-related molecular pathways. The table summarizes main findings in CNS and neurodegeneration correlating LRP8 and its ligand binding to corresponding roles, molecular mechanisms and in vitro and in vivo models used in the experimental studies taken into consideration (see references).
Table 3. LRP8 in CNS- and neurodegeneration-related molecular pathways. The table summarizes main findings in CNS and neurodegeneration correlating LRP8 and its ligand binding to corresponding roles, molecular mechanisms and in vitro and in vivo models used in the experimental studies taken into consideration (see references).
LRP8
Interactors
Roles in
CNS and
Neurodegeneration
LRP8 Main Findings &
Molecular Mechanism
Related to CNS
In Vitro E in Vivo
Models
Refs.
ApoENeurodegenerationIncrease in APP endocytosis
and Aβ production
via X11α or X11β (X11α/β)
Neuroblastoma
N2a cells
[157]
ReelinSynaptic
plasticity
Activation of
synaptic plasticity genes mediated
by the activation
of neuronal enhancers
Primary
cortical neurons/heterozygous Reeler
and LRP8-KO mice
[124]
Enhancement of LRP8
proteolytic processing,
followed by LRP8-ICD
induced transcription
Modulation of NMDA-R
phosphorylation
via SFKs and Dab1 followed by
increased Calcium influx
Primary
wild-type cortical neurons/Dab1
knock-out neurons
[163]
Control of neuronal migration and cellular layer formation in the developing brainPartial inversion
of the neuronal layers
in the neocortex
VLDL and
LRP8 KO mice
[59]
NeurodegenerationActivation of the signaling pathway involving Dab1-PI3K-AKT
leading to the inhibition of GSK3β
and in turn phosphorylation of tau
Primary neurons[161]
Trombospondin-1 (THBS-1)Postnatal neuronal migrationPromotion of
neuroblast chain migration
SVZ explants from wild-type mice, ApoER2−/−
VLDLR−/− mice and THBS-1−/− mice on a C57BL6/J background
[193]
ClusterinPostnatal neuronal migrationModulation of a cell proliferative signal
in migrating neuronal precursors
via Dab1-PI3K/Akt signal
SVZ explants from
wild-type mice
[194]
Selenoprotein P (Sepp1)Preservation of
neurological function and survival
Selenium transportSepp1−/− and Sepp1+/+ male mice
ApoER2−/−
mice (strain name, B6;129S6-Lrp8tm1Her/J)
[199]
F-SpondinNeurodegenerationLRP8 cleavage increase
and Aβ production decrease
COS7 and HEK293 cells transfected with reelin, spondin, thrombospondin
or F-spondin
[158]
SVZ, Subventricular zone.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Passarella, D.; Ciampi, S.; Di Liberto, V.; Zuccarini, M.; Ronci, M.; Medoro, A.; Foderà, E.; Frinchi, M.; Mignogna, D.; Russo, C.; et al. Low-Density Lipoprotein Receptor-Related Protein 8 at the Crossroad between Cancer and Neurodegeneration. Int. J. Mol. Sci. 2022, 23, 8921. https://doi.org/10.3390/ijms23168921

AMA Style

Passarella D, Ciampi S, Di Liberto V, Zuccarini M, Ronci M, Medoro A, Foderà E, Frinchi M, Mignogna D, Russo C, et al. Low-Density Lipoprotein Receptor-Related Protein 8 at the Crossroad between Cancer and Neurodegeneration. International Journal of Molecular Sciences. 2022; 23(16):8921. https://doi.org/10.3390/ijms23168921

Chicago/Turabian Style

Passarella, Daniela, Silvia Ciampi, Valentina Di Liberto, Mariachiara Zuccarini, Maurizio Ronci, Alessandro Medoro, Emanuele Foderà, Monica Frinchi, Donatella Mignogna, Claudio Russo, and et al. 2022. "Low-Density Lipoprotein Receptor-Related Protein 8 at the Crossroad between Cancer and Neurodegeneration" International Journal of Molecular Sciences 23, no. 16: 8921. https://doi.org/10.3390/ijms23168921

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