Skip to main content
  • Research highlight
  • Open access
  • Published:

LRP1: a novel receptor for the transmission of pathological α-Synuclein

During the past decade, mountains of evidence support that the transmission of pathological proteins is a shared mechanism for the progression of various neurodegenerative diseases [1]. α-Synuclein (α-syn) is the pathological protein for a group of neurodegenerative diseases collectively known as α-synucleinopathies, including Parkinson’s disease (PD), dementia with Lewy body (DLB), multiple system atrophy and ~ 50% of Alzheimer’s disease (AD). Blocking the transmission of pathological α-syn represents a promising therapeutic strategy to slow down or reverse the progression of these diseases. Therefore, molecular mechanisms that mediate the transmission of pathological α-syn are attractive drug targets. However, despite extensive study, the molecular mechanisms underlying the transmission of pathological α-syn remains unknown.

Various different mechanisms have been proposed to mediate the transmission of pathological proteins, such as tunneling nanotubes and exosomes [1]. Multiple cell surface receptors have also been reported to bind with pathological proteins and mediate their transmission (Table 1). In 2016, Mao et al. reported that lymphocyte-activation gene 3 (LAG3) receptor mediated the transmission of pathological α-syn in animal and cell models [2]. α-Syn preformed fibrils (PFF) bind directly to the extracellular immunoglobulin-like domains of LAG3. Blocking the interaction between α-syn and LAG3 could reduce the transmission of pathological α-syn. In 2013, Holmes et al. showed in cell models that heparan sulfate proteoglycan (HSPG) mediated macropinocytosis is responsible for the uptake of both pathological α-syn and tau [3]. HSPGs have also been reported to mediate the endocytosis of amyloid-beta (Abeta) [4]. Members of the low-density lipoprotein receptor (LDLR) family have been reported as co-receptors for HSPGs [4, 5]. In 2020, through a functional screen of LDLR receptors, Rauch et al. demonstrated that low-density lipoprotein receptor-related protein 1 (LRP1), is a key receptor that mediated the interneuronal spreading of pathological tau. LRP1 is a member of the LDLR family and expressed in a wide variety of cell types including neurons, astrocytes, microglia, macrophages, fibroblasts, and smooth muscle cells. LRP1 mediates the internalization of physiological tau and pathogenic tau oligomers [6]. LRP1 has also been reported as a co-receptor for HSPG that binds and uptakes Abeta [5]. Moreover, LRP1 has been reported to interact with ApoE, the most significant genetic risk factor for Alzheimer’s disease [7].

Table 1 Receptors mediating the transmission of pathological proteins

Given that HSPGs have been reported to mediate the endocytosis of pathological α-syn, LRP1, as a co-receptor for HSPGs, could also play a critical role in the transmission of pathological α-syn. Published in Molecular Neurodegeneration on September 2, 2022, Chen and colleagues explored the role of the LRP1 in the transmission of α-syn using a wide variety of different models, including LRP1 knockout human induced pluripotent stem cell (iPSc) derived neurons and neuronal LRP1 deletion mice. Loss of LRP1 significantly reduced the internalization of monomeric α-syn, effectively inhibited the uptake of α-syn oligomers, and mildly inhibited the uptake of α-syn PFFs in vitro, suggesting that LRP1 is a mediator for the uptake of soluble α-syn. Using α-syn with modified lysine residues at the N-terminus of the protein, the authors demonstrated that lysine residues and the N-terminus are critical for the interaction and internalization of α-syn. The authors then established a murine model that discriminates between neurons transduced with the adeno-associated virus expressing human α-syn and neurons that receive human α-syn through spreading. Using this novel model, they demonstrated that the deletion of LRP1 in the neurons suppresses the transmission of human α-syn in mice [8].

Illustrating the role of LRP1 in the transmission of α-syn, particularly monomers and oligomers, provides critical insights into the interneuronal transmission mechanism of α-syn, and highlights LRP1 as a promising drug target to block the spreading of oligomeric α-syn in α-synucleinopathies. The usage of various models in the study also dramatically strengthened the conclusion. There are also some questions that remain to be addressed. The very mild effect of LRP1 deletion on α-syn PFF transmission indicates that LRP1 might mediate only a portion of pathological α-syn transmission. Pathological α-syn has been shown to form different conformations, and it remains unknown whether LRP1 plays similar roles in the transmission of various different pathological α-syn conformations. It is also unknown whether post-translational modifications such as phosphorylation on pathological α-syn would modulate the interaction with LRP1. How the existence of co-pathologies such as pathological Abeta and tau would affect LRP1 mediation of pathological α-syn transmission is also unknown. Finally, only the transmission of α-syn monomer has been tested in vivo. Future studies should further explore how LRP1 deletion affects α-syn oligomer and fibril transmission in vivo.

The observation that LRP1 plays a much more important role in the transmission of α-syn monomers and oligomers than fibrils is interesting. The mechanisms, by which LRP1 distinguishes α-syn oligomers versus fibrils remain unknown. Given that α-syn oligomer is a heterogeneous population and the cutoff between fibrils and oligomers is not clearly defined, it would be interesting to further explore the exact size of α-syn oligomers that can be taken up by LRP1 using size exclusion chromatography. Pathological α-syn in diseased brains has been suggested to be a heterogenous population, so the selective uptake of certain pathological species by LRP1 could contribute to selective vulnerability of different brain regions and patients.

Chen and colleagues provide new insights into α-syn transmission. This study highlights LRP1 as a promising drug target for α-synucleinopathies. It also raises many questions that are worth further studies. The exact mechanism of how LRP1 mediates α-syn transmission as well as the interactions between LRP1 and downstream signaling pathways remains to be fully discovered. Evidence from diseased brains will be needed to further support the role of LRP1 in the etiology of α-synucleinopathies. It would also be interesting to explore the role of other LDLRs in the transmission of pathological α-syn. It is possible that different LDLRs could mediate the transmission of different pathological α-syn species.

Availability of data and materials

Not applicable.

Abbreviations

α-syn:

α-Synuclein

PD:

Parkinson's Disease

DLB:

dementia with Lewy body

AD:

Alzheimer's disease

LAG3:

lymphocyte-activation gene 3

PFF:

preformed fibrils

HSPG:

heparan sulfate proteoglycan

Abeta:

amyloid beta

LDLR:

low-density lipoprotein receptor

LRP1:

low-density lipoprotein receptor-related protein 1

iPSc:

induced pluripotent stem cells

References

  1. Peng C, Trojanowski JQ, Lee VM. Protein transmission in neurodegenerative disease. Nat Rev Neurol. 2020;16:199–212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mao X, Ou MT, Karuppagounder SS, Kam TI, Yin X, Xiong Y, et al. Pathological α-synuclein transmission initiated by binding lymphocyte-activation gene 3. Science. 2016;353(6307):Article aah3374.

  3. Holmes BB, DeVos SL, Kfoury N, Li M, Jacks R, Yanamandra K, Ouidja MO, Brodsky FM, Marasa J, Bagchi DP, et al. Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds. Proc Natl Acad Sci USA. 2013;110:E3138–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Shinohara M, Tachibana M, Kanekiyo T, Bu G. Role of LRP1 in the pathogenesis of Alzheimer’s disease: evidence from clinical and preclinical studies. J Lipid Res. 2017;58:1267–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kanekiyo T, Zhang J, Liu Q, Liu CC, Zhang L, Bu G. Heparan sulphate proteoglycan and the low-density lipoprotein receptor-related protein 1 constitute major pathways for neuronal amyloid-beta uptake. J neuroscience: official J Soc Neurosci. 2011;31:1644–51.

    Article  CAS  Google Scholar 

  6. Rauch JN, Luna G, Guzman E, Audouard M, Challis C, Sibih YE, Leshuk C, Hernandez I, Wegmann S, Hyman BT, et al. LRP1 is a master regulator of tau uptake and spread. Nature. 2020;580:381–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kanekiyo T, Bu G. The low-density lipoprotein receptor-related protein 1 and amyloid-beta clearance in Alzheimer’s disease. Front Aging Neurosci. 2014;6:93.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Chen K, Martens YA, Meneses A, Ryu DH, Lu W, Raulin AC, Li F, Zhao J, Chen Y, Jin Y, et al. LRP1 is a neuronal receptor for alpha-synuclein uptake and spread. Mol neurodegeneration. 2022;17:57.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors appreciate the editors for the comments and suggestions.

Funding

This work was supported by NIH/NINDS R01- NS128964, the Cure PSP grant (672-2020-12), and the Michael J. Fox Foundation Grant (MJFF-009049).

Author information

Authors and Affiliations

Authors

Contributions

Crystal Pan wrote and revised the manuscript. Chao Peng designed the manuscript, wrote the manuscript, revised the manuscript, and supervised. The author(s) read and approved the final manuscript. 

Corresponding author

Correspondence to Chao Peng.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they don’t have any competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pan, C., Peng, C. LRP1: a novel receptor for the transmission of pathological α-Synuclein. Mol Neurodegeneration 17, 79 (2022). https://doi.org/10.1186/s13024-022-00582-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13024-022-00582-4

Keywords