1
|
Yang W, Wei Z, Wang T. Unraveling the Role of LRP1 in Alzheimer's Disease: A Focus on Aβ Clearance and the Liver-Brain Axis. J Mol Neurosci 2025; 75:43. [PMID: 40167883 DOI: 10.1007/s12031-025-02339-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, significantly contributing to the global health burden. The progressive accumulation of amyloid-beta (Aβ) plaques and tau tangles triggers neuroinflammation, oxidative stress, and neuronal damage, highlighting the critical need for effective clearance mechanisms. Recent research has identified low-density lipoprotein receptor-related protein 1 (LRP1) as a key factor in the regulation of Aβ clearance, neuroinflammation, and blood-brain barrier integrity, particularly in relation to the liver-brain axis. This review provides a comprehensive examination of the role of LRP1 in AD, focusing on its expression in the brain and liver, its contribution to Aβ metabolism, and its potential as a therapeutic target. Using a systematic literature review, LRP1's multifaceted roles across various biological processes were explored, including its involvement in Aβ transport, clearance via the liver, and modulation of neuroinflammation. Additionally, the impact of physical exercise, pharmacological interventions, and dietary factors on LRP1 expression levels was investigated, elucidating how these approaches may enhance Aβ clearance. The findings demonstrate that LRP1 expression decreases progressively as AD advances, and that augmenting LRP1 activity-particularly through exercise and drug therapies-can improve Aβ clearance and reduce neuroinflammatory responses. Furthermore, LRP1's involvement in the liver-brain axis reveals its broader systemic role in AD pathology. In conclusion, targeting LRP1 offers a promising avenue for AD prevention and treatment, providing new insights into the therapeutic potential of enhancing Aβ clearance pathways through the liver-brain axis.
Collapse
Affiliation(s)
- Wanyue Yang
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China
- Military Medical Sciences Academy, 1 Dali Road, Heping District, Tianjin, 300050, P.R. China
| | - Zilin Wei
- Military Medical Sciences Academy, 1 Dali Road, Heping District, Tianjin, 300050, P.R. China.
| | - Tianhui Wang
- Military Medical Sciences Academy, 1 Dali Road, Heping District, Tianjin, 300050, P.R. China.
| |
Collapse
|
2
|
Majou D, Dermenghem AL. Effects of DHA (omega-3 fatty acid) and estradiol on amyloid β-peptide regulation in the brain. Brain Res 2024; 1823:148681. [PMID: 37992797 DOI: 10.1016/j.brainres.2023.148681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
In the early stages of sporadic Alzheimer's disease (SAD), there is a strong correlation between memory impairment and cortical levels of soluble amyloid-β peptide oligomers (Aβ). It has become clear that Aβ disrupt glutamatergic synaptic function, which can in turn lead to the characteristic cognitive deficits of SAD, but the actual pathways are still not well understood. This opinion article describes the pathogenic mechanisms underlying cerebral amyloidosis. These mechanisms are dependent on the amyloid precursor protein and concern the synthesis of Aβ peptides with competition between the non-amyloidogenic pathway and the amyloidogenic pathway (i.e. a competition between the ADAM10 and BACE1 enzymes), on the one hand, and the various processes of Aβ residue clearance, on the other hand. This clearance mobilizes both endopeptidases (NEP, and IDE) and removal transporters across the blood-brain barrier (LRP1, ABCB1, and RAGE). Lipidated ApoE also plays a major role in all processes. The disturbance of these pathways induces an accumulation of Aβ. The description of the mechanisms reveals two key molecules in particular: (i) free estradiol, which has genomic and non-genomic action, and (ii) free DHA as a preferential ligand of PPARα-RXRα and PPARɣ-RXRα heterodimers. DHA and free estradiol are also self-regulating, and act in synergy. When a certain level of chronic DHA and free estradiol deficiency is reached, a permanent imbalance is established in the central nervous system. The consequences of these deficits are revealed in particular by the presence of Aβ peptide deposits, as well as other markers of the etiology of SAD.
Collapse
Affiliation(s)
- Didier Majou
- ACTIA, 149, rue de Bercy, 75595 Paris Cedex 12, France.
| | | |
Collapse
|
3
|
Rudajev V, Novotny J. Cholesterol-dependent amyloid β production: space for multifarious interactions between amyloid precursor protein, secretases, and cholesterol. Cell Biosci 2023; 13:171. [PMID: 37705117 PMCID: PMC10500844 DOI: 10.1186/s13578-023-01127-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
Amyloid β is considered a key player in the development and progression of Alzheimer's disease (AD). Many studies investigating the effect of statins on lowering cholesterol suggest that there may be a link between cholesterol levels and AD pathology. Since cholesterol is one of the most abundant lipid molecules, especially in brain tissue, it affects most membrane-related processes, including the formation of the most dangerous form of amyloid β, Aβ42. The entire Aβ production system, which includes the amyloid precursor protein (APP), β-secretase, and the complex of γ-secretase, is highly dependent on membrane cholesterol content. Moreover, cholesterol can affect amyloidogenesis in many ways. Cholesterol influences the stability and activity of secretases, but also dictates their partitioning into specific cellular compartments and cholesterol-enriched lipid rafts, where the amyloidogenic machinery is predominantly localized. The most complicated relationships have been found in the interaction between cholesterol and APP, where cholesterol affects not only APP localization but also the precise character of APP dimerization and APP processing by γ-secretase, which is important for the production of Aβ of different lengths. In this review, we describe the intricate web of interdependence between cellular cholesterol levels, cholesterol membrane distribution, and cholesterol-dependent production of Aβ, the major player in AD.
Collapse
Affiliation(s)
- Vladimir Rudajev
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiri Novotny
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
4
|
Israel LL, Sun T, Braubach O, Cox A, Shatalova ES, Rashid HM, Galstyan A, Grodzinski Z, Song XY, Chepurna O, Ljubimov VA, Chiechi A, Sharma S, Phebus C, Wang Y, Ljubimova JY, Black KL, Holler E. β-Amyloid targeting nanodrug for neuron-specific delivery of nucleic acids in Alzheimer's disease mouse models. J Control Release 2023; 361:636-658. [PMID: 37544515 DOI: 10.1016/j.jconrel.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/08/2023]
Abstract
Delivery of therapeutic substances into the brain poses a significant challenge in the treatment of neurological disorders. This is primarily due to the blood-brain barrier (BBB), which restricts access, alongside the limited stability and distribution of these agents within the brain tissue. Here we demonstrate an efficient delivery of microRNA (miRNA) and antisense RNA preferentially to neurons compared to astroglia in the brain of healthy and Alzheimer's disease mice, via disulfide-linked conjugation with poly(ß-L-malic acid-trileucine)-copolymer a biodegradable, amphiphilic, and multivalent platform. By conjugating a D-configured (D3)-peptide (vector) for specific targeting, highly efficient delivery across the BBB is achieved through the Low-Density Lipoprotein Receptor-Related Protein-1 (LRP-1) transcytosis pathway, amyloid beta (Aβ) peptides. Nanodrug distribution was determined by fluorescent labeling and analyzed by microscopy in neurons, astroglia, and in extracellular amyloid plaques typical for Alzheimer's disease. Whereas D-configured BBB-vectors can efficiently target neurons, L-configured (e.g., AP2-peptide) guided vector can only cross BBB but not seem to bind neurons. An analysis of post-injection fluorescence distribution, and RNA-seq followed by real-time PCR validation, confirmed a successful in vivo delivery of morpholino-miRNA-186 nanoconjugates into mouse brain. The size and fluorescence intensity of the intracellular nanodrug particulates were analyzed and verified by a competition with non-fluorescent conjugates. Differentially expressed genes (DEGs) from RNA-seq were identified in the nanodrug injected mice, and the changes of selected DEGs related to Alzheimer's disease were further validated by western blot and real-time PCR. Collectively, these results demonstrated that D3-peptide-conjugated nanopolymer drug is able to achieve neuron-selective delivery of miRNA and can serve as an efficient brain delivery vehicle in Alzheimer's disease (AD) mouse models.
Collapse
Affiliation(s)
- Liron L Israel
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Oliver Braubach
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Alysia Cox
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | | | | | - Anna Galstyan
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Zachary Grodzinski
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Xue Ying Song
- Cedars-Sinai Cancer Applied Genomics Shared Resource, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Oksana Chepurna
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Vladimir A Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Antonella Chiechi
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Sachin Sharma
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Connor Phebus
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Yizhou Wang
- Cedars-Sinai Cancer Applied Genomics Shared Resource, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Julia Y Ljubimova
- Terasaki Institute of Biomedical Innovation, Los Angeles, 90024, USA..
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles 90048, USA.
| | - Eggehard Holler
- Terasaki Institute of Biomedical Innovation, Los Angeles, 90024, USA..
| |
Collapse
|
5
|
Pfundstein G, Nikonenko AG, Sytnyk V. Amyloid precursor protein (APP) and amyloid β (Aβ) interact with cell adhesion molecules: Implications in Alzheimer’s disease and normal physiology. Front Cell Dev Biol 2022; 10:969547. [PMID: 35959488 PMCID: PMC9360506 DOI: 10.3389/fcell.2022.969547] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is an incurable neurodegenerative disorder in which dysfunction and loss of synapses and neurons lead to cognitive impairment and death. Accumulation and aggregation of neurotoxic amyloid-β (Aβ) peptides generated via amyloidogenic processing of amyloid precursor protein (APP) is considered to play a central role in the disease etiology. APP interacts with cell adhesion molecules, which influence the normal physiological functions of APP, its amyloidogenic and non-amyloidogenic processing, and formation of Aβ aggregates. These cell surface glycoproteins also mediate attachment of Aβ to the neuronal cell surface and induce intracellular signaling contributing to Aβ toxicity. In this review, we discuss the current knowledge surrounding the interactions of cell adhesion molecules with APP and Aβ and analyze the evidence of the critical role these proteins play in regulating the processing and physiological function of APP as well as Aβ toxicity. This is a necessary piece of the complex AD puzzle, which we should understand in order to develop safe and effective therapeutic interventions for AD.
Collapse
Affiliation(s)
- Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | | | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
- *Correspondence: Vladimir Sytnyk,
| |
Collapse
|
6
|
Burrinha T, Cláudia GA. Aging impact on amyloid precursor protein neuronal trafficking. Curr Opin Neurobiol 2022; 73:102524. [PMID: 35303572 DOI: 10.1016/j.conb.2022.102524] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 11/03/2022]
Abstract
Neurons live a lifetime. Neuronal aging may increase the risk of Alzheimer's disease. How does neuronal membrane trafficking maintain synapse function during aging? In the normal aged brain, intraneuronal beta-amyloid (Aβ) accumulates without Alzheimer's disease mutations or risk variants. However, do changes with neuronal aging potentiate Aβ accumulation? We reviewed the membrane trafficking of the amyloid precursor protein in neurons and highlighted its importance in Aβ production. Importantly, we reviewed the evidence supporting the impact of aging on neuronal membrane trafficking, APP processing, and consequently Aβ production. Dissecting the molecular regulators of APP trafficking during neuronal aging is required to identify strategies to delay synaptic decline and protect from Alzheimer's disease.
Collapse
Affiliation(s)
- Tatiana Burrinha
- Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal. https://twitter.com/@burrinha_t
| | - Guimas Almeida Cláudia
- Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal.
| |
Collapse
|
7
|
Cuchillo-Ibañez I, Lennol MP, Escamilla S, Mata-Balaguer T, Valverde-Vozmediano L, Lopez-Font I, Ferrer I, Sáez-Valero J. The apolipoprotein receptor LRP3 compromises APP levels. Alzheimers Res Ther 2021; 13:181. [PMID: 34727970 PMCID: PMC8565065 DOI: 10.1186/s13195-021-00921-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Members of the low-density lipoprotein (LDL) receptor family are involved in endocytosis and in transducing signals, but also in amyloid precursor protein (APP) processing and β-amyloid secretion. ApoER2/LRP8 is a member of this family with key roles in synaptic plasticity in the adult brain. ApoER2 is cleaved after the binding of its ligand, the reelin protein, generating an intracellular domain (ApoER2-ICD) that modulates reelin gene transcription itself. We have analyzed whether ApoER2-ICD is able to regulate the expression of other LDL receptors, and we focused on LRP3, the most unknown member of this family. We analyzed LRP3 expression in middle-aged individuals (MA) and in cases with Alzheimer's disease (AD)-related pathology, and the relation of LRP3 with APP. METHODS The effects of full-length ApoER2 and ApoER2-ICD overexpression on protein levels, in the presence of recombinant reelin or Aβ42 peptide, were evaluated by microarray, qRT-PCRs, and western blots in SH-SY5Y cells. LRP3 expression was analyzed in human frontal cortex extracts from MA subjects (mean age 51.8±4.8 years) and AD-related pathology subjects [Braak neurofibrillary tangle stages I-II, 68.4±8.8 years; III-IV, 80.4 ± 8.8 years; V-VI, 76.5±9.7 years] by qRT-PCRs and western blot; LRP3 interaction with other proteins was assessed by immunoprecipitation. In CHO cells overexpressing LRP3, protein levels of full-length APP and fragments were evaluated by western blots. Chloroquine was employed to block the lysosomal/autophagy function. RESULTS We have identified that ApoER2 overexpression increases LRP3 expression, also after reelin stimulation of ApoER2 signaling. The same occurred following ApoER2-ICD overexpression. In extracts from subjects with AD-related pathology, the levels of LRP3 mRNA and protein were lower than those in MA subjects. Interestingly, LRP3 transfection in CHO-PS70 cells induced a decrease of full-length APP levels and APP-CTF, particularly in the membrane fraction. In cell supernatants, levels of APP fragments from the amyloidogenic (sAPPα) or non-amyloidogenic (sAPPβ) pathways, as well as Aβ peptides, were drastically reduced with respect to mock-transfected cells. The inhibitor of lysosomal/autophagy function, chloroquine, significantly increased full-length APP, APP-CTF, and sAPPα levels. CONCLUSIONS ApoER2/reelin signaling regulates LRP3 expression, whose levels are affected in AD; LRP3 is involved in the regulation of APP levels.
Collapse
Affiliation(s)
- Inmaculada Cuchillo-Ibañez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| | - Matthew P Lennol
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Sergio Escamilla
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Trinidad Mata-Balaguer
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Lucía Valverde-Vozmediano
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
| | - Inmaculada Lopez-Font
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Isidro Ferrer
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto de Neuropatología, Hospital Universitario de Bellvitge, Universidad de Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Sáez-Valero
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández de Elche-CSIC, Sant Joan d'Alacant, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.
| |
Collapse
|
8
|
Song XJ, Zhou HY, Sun YY, Huang HC. Phosphorylation and Glycosylation of Amyloid-β Protein Precursor: The Relationship to Trafficking and Cleavage in Alzheimer's Disease. J Alzheimers Dis 2021; 84:937-957. [PMID: 34602469 DOI: 10.3233/jad-210337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder in the central nervous system, and this disease is characterized by extracellular senile plaques and intracellular neurofibrillary tangles. Amyloid-β (Aβ) peptide is the main constituent of senile plaques, and this peptide is derived from the amyloid-β protein precursor (AβPP) through the successive cleaving by β-site AβPP-cleavage enzyme 1 (BACE1) and γ-secretase. AβPP undergoes the progress of post-translational modifications, such as phosphorylation and glycosylation, which might affect the trafficking and the cleavage of AβPP. In the recent years, about 10 phosphorylation sites of AβPP were identified, and they play complex roles in glycosylation modification and cleavage of AβPP. In this article, we introduced the transport and the cleavage pathways of AβPP, then summarized the phosphorylation and glycosylation sites of AβPP, and further discussed the links and relationship between phosphorylation and glycosylation on the pathways of AβPP trafficking and cleavage in order to provide theoretical basis for AD research.
Collapse
Affiliation(s)
- Xi-Jun Song
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China.,Research Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, China
| | - He-Yan Zhou
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China.,Research Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, China
| | - Yu-Ying Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China.,Research Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China.,Research Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, China
| |
Collapse
|
9
|
Analysis of the role of Purα in the pathogenesis of Alzheimer's disease based on RNA-seq and ChIP-seq. Sci Rep 2021; 11:12178. [PMID: 34108502 PMCID: PMC8190037 DOI: 10.1038/s41598-021-90982-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/19/2021] [Indexed: 11/18/2022] Open
Abstract
Purine rich element binding protein A (Purα), encoded by the Purα gene, is an important transcriptional regulator that binds to DNA and RNA and is involved in processes such as DNA replication and RNA translation. Purα also plays an important role in the nervous system. To identify the function of Pura, we performed RNA sequence (RNA-seq) analysis of Purɑ-KO mouse hippocampal neuron cell line (HT22) to analyze the effect of Purα deletion on neuronal expression profiles. And combined with ChIP-seq analysis to explore the mechanism of Purα on gene regulation. In the end, totaly 656 differentially expressed genes between HT22 and Purα-KO HT22 cells have been found, which include 7 Alzheimer’s disease (AD)-related genes and 5 Aβ clearance related genes. 47 genes were regulated by Purα directly, the evidence based on CHIP-seq, which include Insr, Mapt, Vldlr, Jag1, etc. Our study provides the important informations of Purα in neuro-development. The possible regulative effects of Purα on AD-related genes consist inthe direct and indirect pathways of Purα in the pathogenesis of AD.
Collapse
|
10
|
Trafficking in Alzheimer's Disease: Modulation of APP Transport and Processing by the Transmembrane Proteins LRP1, SorLA, SorCS1c, Sortilin, and Calsyntenin. Mol Neurobiol 2017; 55:5809-5829. [PMID: 29079999 DOI: 10.1007/s12035-017-0806-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/17/2017] [Indexed: 12/11/2022]
Abstract
The amyloid precursor protein (APP), one key player in Alzheimer's disease (AD), is extensively processed by different proteases. This leads to the generation of diverging fragments including the amyloid β (Aβ) peptide, which accumulates in brains of AD patients. Subcellular trafficking of APP is an important aspect for its proteolytic conversion, since the various secretases which cleave APP are located in different cellular compartments. As a consequence, altered subcellular targeting of APP is thought to directly affect the degree to which Aβ is generated. The mechanisms underlying intracellular APP transport are critical to understand AD pathogenesis and can serve as a target for future pharmacological interventions. In the recent years, a number of APP interacting proteins were identified which are implicated in sorting of APP, thereby influencing APP processing at different angles of the secretory or endocytic pathway. This review provides an update on the proteolytic processing of APP and the interplay of the transmembrane proteins low-density lipoprotein receptor-related protein 1, sortilin-receptor with A-type repeats, SorCS1c, sortilin, and calsyntenin. We discuss the specific interactions with APP, the capacity to modulate the intracellular itinerary and the proteolytic conversion of APP, a possible involvement in the clearance of Aβ, and the implications of these transmembrane proteins in AD and other neurodegenerative diseases.
Collapse
|
11
|
Storck SE, Pietrzik CU. Endothelial LRP1 - A Potential Target for the Treatment of Alzheimer's Disease : Theme: Drug Discovery, Development and Delivery in Alzheimer's Disease Guest Editor: Davide Brambilla. Pharm Res 2017; 34:2637-2651. [PMID: 28948494 DOI: 10.1007/s11095-017-2267-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/15/2017] [Indexed: 12/19/2022]
Abstract
The accumulation of the neurotoxin beta-amyloid (Aβ) is a major hallmark in Alzheimer's disease (AD). Aβ homeostasis in the brain is governed by its production and various clearance mechanisms. Both pathways are influenced by the ubiquitously expressed low-density lipoprotein receptor-related protein 1 (LRP1). In cerebral blood vessels, LRP1 is an important mediator for the rapid removal of Aβ from brain via transport across the blood-brain barrier (BBB). Here, we summarize recent findings on LRP1 function and discuss the targeting of LRP1 as a modulator for AD pathology and drug delivery into the brain.
Collapse
Affiliation(s)
- Steffen E Storck
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Duesbergweg 6, 55099, Mainz, Germany
| | - Claus U Pietrzik
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Duesbergweg 6, 55099, Mainz, Germany.
| |
Collapse
|