Skip to main content
BMB Reports logoLink to BMB Reports
. 2023 Nov 14;56(12):645–650. doi: 10.5483/BMBRep.2023-0151

The role of cellular prion protein in immune system

Seunghwa Cha 1, Mi-Yeon Kim 1,*
PMCID: PMC10761747  PMID: 37817440

Abstract

Numerous studies have investigated the cellular prion protein (PrPC) since its discovery. These investigations have explained that its structure is predominantly composed of alpha helices and short beta sheet segments, and when its abnormal scrapie isoform (PrPSc) is infected, PrPSc transforms the PrPC, leading to prion diseases, including Creutzfeldt-Jakob disease in humans and bovine spongiform encephalopathy in cattle. Given its ubiquitous distribution across a variety of cellular types, the PrPC manifests a diverse range of biological functions, including cell-cell adhesion, neuroprotection, signalings, and oxidative stress response. PrPC is also expressed in immune tissues, and its functions in these tissues include the activation of immune cells and the formation of secondary lymphoid tissues, such as the spleen and lymph nodes. Moreover, high expression of PrPC in immune cells plays a crucial role in the pathogenesis of prion diseases. In addition, it affects inflammation and the development and progression of cancer via various mechanisms. In this review, we discuss the studies on the role of PrPC from various immunological perspectives.

Keywords: Cancer, Immune system, Inflammation, Prion, Prion disease

INTRODUCTION

The cellular prion protein (PrPC) is predominantly found in the brain tissue and neurons, where it is attached to the cell membrane (1-3). PrPC is a single polypeptide composed of 208 amino acids in mice and 209 amino acids in humans (4-6). In the human PrPC, there are two N-glycosylation sites located at residue 181 and 197. Through Western blotting, diglycosylated (36 kDa), monoglycosylated (33 kDa), and unglycosylated cerebral PrP isoforms (27 kDa) can be distinguished (7, 8). PrPC exhibits a predominantly alpha-helical structure with a flexible N-terminal domain and a globular C-terminal domain. The C-terminal region contains three alpha helices and two short segments of the beta sheet structure. For its abnormal scrapie isoform (PrPSc), PrPC undergoes a conformational change that converts alpha helices into beta-sheets (6, 9, 10). This conformational alteration is associated with the pathogenicity and transmission of prion diseases (3, 11-13). Prion diseases, also known as transmissible spongiform encephalopathies, are rare and fatal neurodegenerative disorders that are caused by the accumulation of abnormally folded PrPSc in the brain of humans and animals (14-16). The abnormal PrPSc form acts as a template and induces the conversion of PrPC into a disease-associated form. This results in progressive accumulation of PrPSc, which disrupts normal brain function and causes neuronal damage (17-19). Based on the specific type and region of the brain affected, prion diseases manifest with different symptoms, including cognitive impairment, memory loss, behavioral changes, movement disorders, and severe neurological dysfunction (20, 21). Several prion diseases affect humans, including Creutzfeldt-Jakob disease (CJD), variant CJD, Gerstmann-Sträussler-Scheinker syndrome, and fatal familial insomnia (22, 23). Each disease has distinct clinical and pathological features.

PrPC plays multifaceted roles in various cellular processes, including cell-cell adhesion (24), neuroprotection (25, 26), intracellular signaling (27), cell death and survival (28), and oxidative stress response (29). In addition to nerve cells, PrPC is also expressed in various other cells, especially leukocytes, which are a part of the immune system (30). Despite the high expression of PrPC in leukocytes, there have been limited scientific investigations of its precise role of PrPC.

Here, we provide an overview of the studies investigating the role of PrPC in the immune system. Specifically, we discuss the expression of PrPC in immune cells and its function in the structural formation of secondary lymphoid tissues, such as the spleen and lymph nodes. Furthermore, we focused on the mechanisms of peripheral PrPSc replication and neuroinvasion of PrPSc via the immune system. We also examined the involvement of PrPC in inflammation, cancer development, and metastasis.

EXPRESSION OF PrPC ON IMMUNE CELLS

PrPC is widely expressed on cell surfaces at different levels in immune cells, such as T lymphocytes (31, 32), natural killer (NK) cells (33, 34), macrophages (35), dendritic cells (DCs) (36), regulatory T cells (37) and follicular dendritic cells (FDCs) (38). Although the role of PrPC in immune cells is not well understood, PrPC expression increases during NK cell differentiation and functional maturation (33, 34). In addition, PrPC in human T cells interacts with the transducer protein zeta-chain associated protein-70, which plays a critical role in the signaling pathway leading to T cell activation (31). Elevated levels of PrPC expression have been found to facilitate T lymphocyte activation, promote cell proliferation, and enhance differentiation through the T cell receptor signaling pathway (32). DCs also exhibit upregulation of PrPC expression after maturation (36). PrPC also affects the phagocytic capacity of macrophages by activating the ERK1/2 and Akt kinases (35). Taken together, these studies demonstrate a correlation between the activation of immune cells and PrPC expression, although the exact role of upregulated PrPC remains uncertain.

Regulatory T cells, known for their ability to suppress immune responses, express higher levels of PrPC than conventional T cells (37). Our recent study showed that PrPC is involved in the development and function of regulatory T cells and it will be discussed in detail in section ‘Role of regulatory T cells in cancer and their relationship with PrPC.’

FDCs are a specialized type of non-hematopoietic immune cell found primarily within the B cell area of secondary lymphoid tissues and are recognized for their high expression of PrPC. During infection with PrPSc, they serve as the initial sites of accumulation in the lymphoid tissues before PrPSc subsequently spreads to the central nervous system (CNS) (38). However, specific ablation of PrPC expression in FDCs using Cre-mediated recombination did not affect the normal function of FDCs (39). Further studies are required to elucidate the functions of PrPC in FDCs.

INVOLVEMENT OF PrPC IN THE STRUCTURE OF SECONDARY LYMPHOID TISSUES

To investigate the role of PrPC in the structure and organization of secondary lymphoid tissues, such as the spleen and lymph nodes, several studies have been performed using mice lacking PrPC (Prnp0/0) and mice infected with the mouse-adapted scrapie strain ME7 (40-42). These studies revealed that PrPC plays an important role in the formation and maintenance of secondary lymphoid tissue structures. Spleen obtained from Prnp0/0 and ME7-infected mice showed impaired structures, with a lack of segregation between the T and B zones in the white pulp region. In both cases, there was no or significant reduction in the size of the T-zone. This can be attributed to the decreased expression of T-cell homing chemokines CCL19 and CCL21, which are involved in T-zone formation (43), leading to impaired recruitment of CD4 T cells in both mouse models (40, 41). Although both Prnp0/0 and ME7-infected spleens exhibited impaired T-zone structures, when compared to uninfected wild-type mice, the number of lymphoid tissue inducer (LTi) cells, which are important for secondary lymphoid tissue development (44, 45) decreased in Prnp0/0 spleens but remained unchanged in ME7-infected spleens. These results suggest that persistent PrPC, without conversion to PrPSc in ME7-infected mice likely regulates both the quantity of LTi cells and their migration to the spleen.

PATHOGENESIS OF PRION DISEASES VIA IMMUNE SYSTEM

Prion diseases develop when PrPSc infects the host. Infected PrPSc continuously converts PrPC to PrPSc, leading to the accumulation of PrPSc. Ultimately, the accumulation of PrPSc leads to the onset of prion diseases (46, 47). PrPSc accumulation primarily occurs in the immune system as immune cells express PrPC (47-49). Following infection, PrPSc circulates through the bloodstream and reaches secondary lymphoid tissues such as the lymph nodes, tonsils, Peyer’s patches, and spleen. At these sites, PrPSc utilizes immune cells to replicate and accumulate (46, 50). Once sufficient replication and accumulation occur, prion diseases are triggered subsequent to CNS infection, initiating the progression of pathological manifestations. Therefore, PrPSc accumulation in the secondary lymphoid tissues is crucial for movement of PrPSc into the CNS.

FDCs found in the B cell area of germinal centers (GCs) in secondary lymphoid tissues are critical for capturing naïve antigens using FcγRIIB and complement receptors and presenting them to GC B cells (51-53). In response to these antigens, B cells receive help from follicular helper T (Tfh) cells, which leads to their activation and subsequent differentiation into plasma cells (54-56). Due to high PrPC expression levels, FDCs accumulate considerable amount of PrPSc upon infection. Consequently, FDCs are pivotal in the onset of prion diseases, and the spleen serves as the primary site for PrPSc replication mediated by FDCs (57-60). In the absence of FDCs, the lack of a site for accumulation of PrPSc prevents its accumulation. Consequently, neuroinvasion does not occur in the absence of PrPSc accumulation (60-62).

Our study showed that ME7-infected mice exhibited increased FDC networks and Tfh cell responses, which persisted throughout the progression of prion disease (42). Despite a decrease in CD4 T cells in the white pulp, there was an increase in CD4 T cells within GCs, accompanied by higher expression levels of Tfh-related genes, such as Bcl6, Il21, Cxcr5, Icos, and Pdcd1. Moreover, the ME7-infected spleens showed an elevated number of CD4 memory T cells. These results suggest that although ME7 infection led to an impaired structure in the splenic white pulp, there was an expansion of CD4 memory T cells and prolonged Tfh cell responses necessary to support the replication and accumulation of PrPSc within GCs.

ROLE OF PrPC IN INFLAMMATION

Several studies have investigated the effects of PrPC on inflammatory responses, given its substantial expression in immune cells and its ability to influence immune cell activation and recruitment. These studies revealed that PrPC protects organs from inflammatory responses through its immunomodulatory function (63-67). High expression of PrPC in immune-privileged organs, including the brain, placenta, eyes, testes, and uterus, serves as a protective mechanism against inflammation-induced damage to these organs (63). Inflammation studies using Prnp-knockout models have been conducted to investigate the protective role of PrPC against inflammation (68-71). Tsutsui et al. demonstrated that Prnp0/0 mice immunized with myelin oligodendrocyte glycoprotein peptide to induce experimental autoimmune encephalomyelitis exhibited a more aggressive disease onset characterized by higher levels of leukocytic infiltrates and increased expression of pro-inflammatory cytokine genes in the brain and spinal cord suggesting the protective role of PrPC against neuroinflammation (68). Petit et al. investigated the severity of inflammatory bowel disease induced by dextran sodium sulfate and found that mice lacking PrPC exhibited more severe symptoms than wild-type mice (69). Furthermore, when PrPC was knocked down in human enterocytes, there was a decrease in cell-cell junctions. The weakening of the intestinal barrier, consisting of tight cell-cell junctions, can lead to vulnerability to external invasion and infection, potentially resulting in inflammatory bowel diseases such as Crohn’s disease and ulcerative colitis. Consistent with these findings, patients with Crohn’s disease or ulcerative colitis showed decreased levels of PrPC at cell-cell junctions in the colonic epithelia (69). Taken together, these results revealed that in the absence of PrPC, there was an increase in inflammatory responses and a greater extent of associated damage.

In another study, goats lacking PrPC exhibited prolonged symptoms in response to lipopolysaccharide-induced (LPS)-induced systemic inflammation (70). In another study using LPS, Liu et al. showed that upon LPS injection, wild-type mice exhibited elevated levels of pro-inflammatory cytokines in the brain and spleen during the acute phase, whereas Prnp0/0 mice displayed lower cytokine levels (71). Additionally, Prnp0/0 mice showed higher mortality rates in response to LPS-induced septic shock. These results suggest that PrPC plays a crucial role in protecting against LPS injection by modulating the inflammatory response.

ROLE OF PrPC IN CANCER DEVELOPMENT AND METASTASIS

Several studies have demonstrated that PrPC stimulates cancer cell proliferation through various mechanisms. Elevated levels of PrPC expression have been linked to unfavorable prognoses and have been observed in various human cancers such as gastric carcinoma (72), renal adenocarcinoma (73), colorectal cancer (74, 75), breast cancer (76, 77), and melanoma (78). In gastric cancer cells, PrPC overexpression activates the phosphatidylinositide 3-kinase pathway and upregulation of cyclin D1, promoting the G1/S phase transition and consequently facilitating cell proliferation (79). Another study reported that PrPC influences the G1/S phase transition in several renal adenocarcinoma cell lines (80). Consistent with these findings, in the absence of PrPC, the expression of cyclins and cyclin-dependent kinases is suppressed, leading to the inhibition of cell proliferation in colon cancer (81).

Gil et al. demonstrated that PrPC contributes to the invasion and migration of breast cancer cells by regulating matrix metalloprotease-9 (MMP-9) (82). They showed that overexpression of PrPC in the breast cancer cell line MCF-7 leads to an increase in MMP-9 expression by enhancing the association of NF-κB with the promoter region of the MMP-9 gene and activating the ERK signaling pathway. Conversely, when PrPC is silenced using siRNA, a notable decrease in ERK activation and MMP-9 expression is observed, leading to the suppression of cell migration and invasion (82).

Reportedly, PrPC plays a role in both cancer development and metastasis, the process by which cancer cells spread from the primary tumor to other parts of the body (72, 83, 84). Metastatic gastric cancers exhibit high PrPC expression, which plays a substantial role in enhancing the adhesive, invasive, and metastatic capacities of gastric cancer cell lines (72). According to Pan et al., the N-terminal region of PrPC can activate the MEK/ERK pathway, ultimately leading to transactivation of MMP11. This activation enhances the invasive and metastatic properties of gastric cancer cells, indicating their potential role in promoting metastasis (72). Additionally, the overexpression of MMP11 is frequently associated with a more aggressive tumor phenotype and resistance to apoptosis (83). Wang et al. showed that PrPC is specifically expressed at the invasive front of colorectal cancers (CRCs), promoting tumor invasion through the acquisition of characteristics associated with epithelial-mesenchymal transition (84). Additionally, they showed that knockdown of PrPC in an orthotopic xenograft model significantly reduced the number of distant metastases, supporting the significant role of PrPC in the regulation of CRC progression and metastasis.

ROLE OF REGULATORY T CELLS IN CANCER AND THEIR RELATIONSHIP WITH PrPC

Research on the role of PrPC in cancer invasion and metastasis has revealed its association with regulatory T cells (37, 78). Regulatory T cells possess immunosuppressive functions and exhibit elevated activity in numerous types of cancers (85, 86). An increased number of tumor-infiltrating regulatory T cells have been identified, and their increased activity are observed in various human cancers, including liver, lung, breast, gastrointestinal tract, pancreas, and ovarian cancers (85-87). Our recent study showed that when B16F10 melanoma cells were injected into Prnp0/0 and PrP-overexpressing (Tga20) mice to induce lung metastasis, Tga20 mice reached the terminal stage much faster than Prnp0/0 mice as lung metastasis occurred (78). This effect was likely associated with an increased number of regulatory T cells. The expression of transforming growth factor-beta (TGF-β) and programmed death ligand-1 (PD-L1), which play important roles in the differentiation and function of regulatory T cells, was upregulated in Tga20 mice compared with Prnp0/0 mice. These results suggest that during the invasion and metastasis of cancer cells, PrPC may contribute to the development and activation of regulatory T cells by increasing the expression of TGF-β and PD-L1, which enhances their functionality.

CONCLUSION

In almost all cells, PrPC is expressed, participating in diverse cellular processes and also found in immune cells, suggesting potential roles in the immune system. Based on the research findings to date, it has been revealed that PrPC has a protective function in inflammatory responses triggered by infections, but contributes to the development of cancer through various mechanisms, including regulation of cell proliferation, enhancement of adhesive, invasive, and metastatic capacities, and boosting regulatory T cell activation. These findings suggest that PrPC plays a crucial role in the immune system and has substantial implications in the pathogenesis of conditions like inflammation and cancer. However, since much of this research is based on results of phenomena observed through knockout systems, further investigations are required to unveil the specific roles of PrPC in individual immune cells, elucidate molecular mechanisms, and understand the interactions through which PrPC influences the immune system. Future studies exploring the precise functions of PrPC in the immune system will enable the regulation of the immune system using PrPC, which can be applied for the treatment of various diseases.

Funding Statement

ACKNOWLEDGEMENTS This work was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Education, Science, and Technology (NRF- 2016R1D1A1B01011371).

Footnotes

CONFLICTS OF INTEREST

The authors have no conflicting interests.

REFERENCES

  • 1.Mabbott NA, Brown KL, Manson J, Bruce ME. T-lymphocyte activation and the cellular form of the prion protein. Immunology. 1997;92:161–165. doi: 10.1046/j.1365-2567.1997.00331.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Antoine N, Cesbron JY, Coumans B, Jolois O, Zorzi W, Heinen E. Differential expression of cellular prion protein on human blood and tonsil lymphocytes. Haematologica. 2000;85:475–480. [PubMed] [Google Scholar]
  • 3.Stahl N, Borchelt DR, Hsiao K, Prusiner SB. Scrapie prion protein contains a phosphatidylinositol glycolipid. Cell. 1987;51:229–240. doi: 10.1016/0092-8674(87)90150-4. [DOI] [PubMed] [Google Scholar]
  • 4.Riek R, Hornemann S, Wider G, Glockshuber R, Wuthrich K. NMR characterization of the full-length recombinant murine prion protein, mPrP(23-231) FEBS Lett. 1997;413:282–288. doi: 10.1016/S0014-5793(97)00920-4. [DOI] [PubMed] [Google Scholar]
  • 5.Sparkes RS, Simon M, Cohn VH, et al. Assignment of the human and mouse prion protein genes to homologous chromosomes. Proc Natl Acad Sci U S A. 1986;83:7358–7362. doi: 10.1073/pnas.83.19.7358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zahn R, Liu A, Luhrs T, et al. NMR solution structure of the human prion protein. Proc Natl Acad Sci U S A. 2000;97:145–150. doi: 10.1073/pnas.97.1.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Schmitz M, Lullmann K, Zafar S, et al. Association of prion protein genotype and scrapie prion protein type with cellular prion protein charge isoform profiles in cerebrospinal fluid of humans with sporadic or familial prion diseases. Neurobiol Aging. 2014;35:1177–1188. doi: 10.1016/j.neurobiolaging.2013.11.010. [DOI] [PubMed] [Google Scholar]
  • 8.Yang Y, Chen L, Pan HZ, Kou Y, Xu CM. Glycosylation modification of human prion protein provokes apoptosis in HeLa cells in vitro. BMB Rep. 2009;42:331–337. doi: 10.5483/BMBRep.2009.42.6.331. [DOI] [PubMed] [Google Scholar]
  • 9.Damberger FF, Christen B, Perez DR, Hornemann S, Wuthrich K. Cellular prion protein conformation and function. Proc Natl Acad Sci U S A. 2011;108:17308–17313. doi: 10.1073/pnas.1106325108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Riesner D. Biochemistry and structure of PrP(C) and PrP(Sc) Br Med Bull. 2003;66:21–33. doi: 10.1093/bmb/66.1.21. [DOI] [PubMed] [Google Scholar]
  • 11.Pan KM, Baldwin M, Nguyen J, et al. Conversion of alpha-helices into beta-sheets features in the formation of the scrapie prion proteins. Proc Natl Acad Sci U S A. 1993;90:10962–10966. doi: 10.1073/pnas.90.23.10962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wille H, Requena JR. The Structure of PrP(Sc) Prions. Pathogens. 2018;7:20. doi: 10.3390/pathogens7010020.6a46b21943524c4ab0dfb43c76af37e0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Diaz-Espinoza R, Soto C. High-resolution structure of infectious prion protein: the final frontier. Nat Struct Mol Biol. 2012;19:370–377. doi: 10.1038/nsmb.2266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chesebro B. Introduction to the transmissible spongiform encephalopathies or prion diseases. Br Med Bull. 2003;66:1–20. doi: 10.1093/bmb/66.1.1. [DOI] [PubMed] [Google Scholar]
  • 15.Peggion C, Bertoli A, Sorgato MC. Almost a century of prion protein(s): from pathology to physiology, and back to pathology. Biochem Biophys Res Commun. 2017;483:1148–1155. doi: 10.1016/j.bbrc.2016.07.118. [DOI] [PubMed] [Google Scholar]
  • 16.Chen RJ, Chang WW, Lin YC, Cheng PL, Chen YR. Alzheimer's amyloid-beta oligomers rescue cellular prion protein induced tau reduction via the Fyn pathway. ACS Chem Neurosci. 2013;4:1287–1296. doi: 10.1021/cn400085q. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nicotera P. A route for prion neuroinvasion. Neuron. 2001;31:345–348. doi: 10.1016/S0896-6273(01)00385-3. [DOI] [PubMed] [Google Scholar]
  • 18.Daude N. Prion diseases and the spleen. Viral Immunol. 2004;17:334–349. doi: 10.1089/vim.2004.17.334. [DOI] [PubMed] [Google Scholar]
  • 19.Wierzbicka A, Deptula W. The role of the immune system in the pathogenesis of prion diseases. Postepy Hig Med Dosw 62. 2008;(Online):166–173. [PubMed] [Google Scholar]
  • 20.Thompson A, MacKay A, Rudge P, et al. Behavioral and psychiatric symptoms in prion disease. Am J Psychiatry. 2014;171:265–274. doi: 10.1176/appi.ajp.2013.12111460. [DOI] [PubMed] [Google Scholar]
  • 21.Kim HO, Snyder GP, Blazey TM, Race RE, Chesebro B, Skinner PJ. Prion disease induced alterations in gene expression in spleen and brain prior to clinical symptoms. Adv Appl Bioinform Chem. 2008;1:29–50. doi: 10.2147/AABC.S3411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Budka H, Aguzzi A, Brown P, et al. Neuropathological diagnostic criteria for Creutzfeldt-Jakob disease (CJD) and other human spongiform encephalopathies (prion diseases) Brain Pathol. 1995;5:459–466. doi: 10.1111/j.1750-3639.1995.tb00625.x. [DOI] [PubMed] [Google Scholar]
  • 23.Kitamoto T, Muramoto T, Mohri S, Doh-Ura K, Tateishi J. Abnormal isoform of prion protein accumulates in follicular dendritic cells in mice with Creutzfeldt-Jakob disease. J Virol. 1991;65:6292–6295. doi: 10.1128/jvi.65.11.6292-6295.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Malaga-Trillo E, Solis GP, Schrock Y, et al. Regulation of embryonic cell adhesion by the prion protein. PLoS Biol. 2009;7:e55. doi: 10.1371/journal.pbio.1000055.a9a69c6eb6874bedb300ea793e98bf61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lopes MH, Hajj GN, Muras AG, et al. Interaction of cellular prion and stress-inducible protein 1 promotes neuritogenesis and neuroprotection by distinct signaling pathways. J Neurosci. 2005;25:11330–11339. doi: 10.1523/JNEUROSCI.2313-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Doeppner TR, Kaltwasser B, Schlechter J, et al. Cellular prion protein promotes post-ischemic neuronal survival, angioneurogenesis and enhances neural progenitor cell homing via proteasome inhibition. Cell Death Dis. 2015;6:e2024. doi: 10.1038/cddis.2015.365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lee YJ, Baskakov IV. The cellular form of the prion protein guides the differentiation of human embryonic stem cells into neuron-, oligodendrocyte-, and astrocyte-committed lineages. Prion. 2014;8:266–275. doi: 10.4161/pri.32079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bravard A, Auvre F, Fantini D, et al. The prion protein is critical for DNA repair and cell survival after genotoxic stress. Nucleic Acids Res. 2015;43:904–916. doi: 10.1093/nar/gku1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kouadri A, El Khatib M, Cormenier J, et al. Involvement of the prion protein in the protection of the human bronchial epithelial barrier against oxidative stress. Antioxid Redox Signal. 2019;31:59–74. doi: 10.1089/ars.2018.7500. [DOI] [PubMed] [Google Scholar]
  • 30.Isaacs JD, Jackson GS, Altmann DM. The role of the cellular prion protein in the immune system. Clin Exp Immunol. 2006;146:1–8. doi: 10.1111/j.1365-2249.2006.03194.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Mattei V, Garofalo T, Misasi R, et al. Prion protein is a component of the multimolecular signaling complex involved in T cell activation. FEBS Lett. 2004;560:14–18. doi: 10.1016/S0014-5793(04)00029-8. [DOI] [PubMed] [Google Scholar]
  • 32.Li R, Liu D, Zanusso G, et al. The expression and potential function of cellular prion protein in human lymphocytes. Cell Immunol. 2001;207:49–58. doi: 10.1006/cimm.2000.1751. [DOI] [PubMed] [Google Scholar]
  • 33.Durig J, Giese A, Schulz-Schaeffer W, et al. Differential constitutive and activation-dependent expression of prion protein in human peripheral blood leucocytes. Br J Haematol. 2000;108:488–495. doi: 10.1046/j.1365-2141.2000.01881.x. [DOI] [PubMed] [Google Scholar]
  • 34.Dodelet VC, Cashman NR. Prion protein expression in human leukocyte differentiation. Blood. 1998;91:1556–1561. doi: 10.1182/blood.V91.5.1556. [DOI] [PubMed] [Google Scholar]
  • 35.Krebs B, Dorner-Ciossek C, Schmalzbauer R, Vassallo N, Herms J, Kretzschmar HA. Prion protein induced signaling cascades in monocytes. Biochem Biophys Res Commun. 2006;340:13–22. doi: 10.1016/j.bbrc.2005.11.158. [DOI] [PubMed] [Google Scholar]
  • 36.Martinez del Hoyo G, Lopez-Bravo M, Metharom P, Ardavin C, Aucouturier P. Prion protein expression by mouse dendritic cells is restricted to the nonplasmacytoid subsets and correlates with the maturation state. J Immunol. 2006;177:6137–6142. doi: 10.4049/jimmunol.177.9.6137. [DOI] [PubMed] [Google Scholar]
  • 37.Isaacs JD, Garden OA, Kaur G, Collinge J, Jackson GS, Altmann DM. The cellular prion protein is preferentially expressed by CD4+ CD25+ Foxp3+ regulatory T cells. Immunology. 2008;125:313–319. doi: 10.1111/j.1365-2567.2008.02853.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.McCulloch L, Brown KL, Bradford BM, et al. Follicular dendritic cell-specific prion protein (PrP) expression alone is sufficient to sustain prion infection in the spleen. PLoS Pathog. 2011;7:e1002402. doi: 10.1371/journal.ppat.1002402.4907cbe0c0c0411d8a32bde29ccc06df [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.McCulloch L, Brown KL, Mabbott NA. Ablation of the cellular prion protein, PrPC, specifically on follicular dendritic cells has no effect on their maturation or function. Immunology. 2013;138:246–257. doi: 10.1111/imm.12031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kim S, Han S, Lee YE, et al. Prion protein-deficient mice exhibit decreased CD4 T and LTi cell numbers and impaired spleen structure. Immunobiology. 2016;221:94–102. doi: 10.1016/j.imbio.2015.07.017. [DOI] [PubMed] [Google Scholar]
  • 41.Kim S, Han S, Lee HS, Kim YS, Choi EK, Kim MY. Impaired spleen structure and chemokine expression in ME7 scrapie-infected mice. Immunobiology. 2016;221:871–878. doi: 10.1016/j.imbio.2016.03.008. [DOI] [PubMed] [Google Scholar]
  • 42.Kim S, Han S, Kim T, et al. Prolonged follicular helper T cell responses in ME7 scrapie-infected mice. Prion. 2018;12:109–116. doi: 10.1080/19336896.2018.1458573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Fevang B, Yndestad A, Damas JK, et al. Chemokines and common variable immunodeficiency; possible contribution of CCL19, CCL21 and CCR7 to immune dysregulation. Clin Exp Immunol. 2009;158:237–245. doi: 10.1111/j.1365-2249.2009.04013.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kim MY, Gaspal FM, Wiggett HE, et al. CD4(+)CD3(−) accessory cells costimulate primed CD4 T cells through OX40 and CD30 at sites where T cells collaborate with B cells. Immunity. 2003;18:643–654. doi: 10.1016/S1074-7613(03)00110-9. [DOI] [PubMed] [Google Scholar]
  • 45.Kim MY, Toellner KM, White A, et al. Neonatal and adult CD4+ CD3- cells share similar gene expression profile, and neonatal cells up-regulate OX40 ligand in response to TL1A (TNFSF15) J Immunol. 2006;177:3074–3081. doi: 10.4049/jimmunol.177.5.3074. [DOI] [PubMed] [Google Scholar]
  • 46.Fraser H, Dickinson AG. Pathogenesis of scrapie in the mouse: the role of the spleen. Nature. 1970;226:462–463. doi: 10.1038/226462a0. [DOI] [PubMed] [Google Scholar]
  • 47.Kimberlin RH, Walker CA. Pathogenesis of mouse scrapie: evidence for neural spread of infection to the CNS. J Gen Virol. 1980;51:183–187. doi: 10.1099/0022-1317-51-1-183. [DOI] [PubMed] [Google Scholar]
  • 48.Zhang B, Shen P, Yin X, Dai Y, Ding M, Cui L. Expression and functions of cellular prion proteins in immunocytes. Scand J Immunol. 2020;91:e12854. doi: 10.1111/sji.12854. [DOI] [PubMed] [Google Scholar]
  • 49.Brandner S. CNS pathogenesis of prion diseases. Br Med Bull. 2003;66:131–139. doi: 10.1093/bmb/66.1.131. [DOI] [PubMed] [Google Scholar]
  • 50.Fraser H, Dickinson AG. Studies of the lymphoreticular system in the pathogenesis of scrapie: the role of spleen and thymus. J Comp Pathol. 1978;88:563–573. doi: 10.1016/0021-9975(78)90010-5. [DOI] [PubMed] [Google Scholar]
  • 51.Liu YJ, Grouard G, de Bouteiller O, Banchereau J. Follicular dendritic cells and germinal centers. Int Rev Cytol. 1996;166:139–179. doi: 10.1016/S0074-7696(08)62508-5. [DOI] [PubMed] [Google Scholar]
  • 52.Aguzzi A, Kranich J, Krautler NJ. Follicular dendritic cells: origin, phenotype, and function in health and disease. Trends Immunol. 2014;35:105–113. doi: 10.1016/j.it.2013.11.001. [DOI] [PubMed] [Google Scholar]
  • 53.Heesters BA, Myers RC, Carroll MC. Follicular dendritic cells: dynamic antigen libraries. Nat Rev Immunol. 2014;14:495–504. doi: 10.1038/nri3689. [DOI] [PubMed] [Google Scholar]
  • 54.Fischer MB, Goerg S, Shen L, et al. Dependence of germinal center B cells on expression of CD21/CD35 for survival. Science. 1998;280:582–585. doi: 10.1126/science.280.5363.582. [DOI] [PubMed] [Google Scholar]
  • 55.Gitlin AD, Mayer CT, Oliveira TY, et al. HUMORAL IMMUNITY. T cell help controls the speed of the cell cycle in germinal center B cells. Science. 2015;349:643–646. doi: 10.1126/science.aac4919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zotos D, Coquet JM, Zhang Y, et al. IL-21 regulates germinal center B cell differentiation and proliferation through a B cell-intrinsic mechanism. J Exp Med. 2010;207:365–378. doi: 10.1084/jem.20091777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.McGovern G, Brown KL, Bruce ME, Jeffrey M. Murine scrapie infection causes an abnormal germinal centre reaction in the spleen. J Comp Pathol. 2004;130:181–194. doi: 10.1016/j.jcpa.2003.11.001. [DOI] [PubMed] [Google Scholar]
  • 58.Brown KL, Stewart K, Ritchie DL, et al. Scrapie replication in lymphoid tissues depends on prion protein-expressing follicular dendritic cells. Nat Med. 1999;5:1308–1312. doi: 10.1038/15264. [DOI] [PubMed] [Google Scholar]
  • 59.Raeber AJ, Montrasio F, Hegyi I, et al. Studies on prion replication in spleen. Dev Immunol. 2001;8:291–304. doi: 10.1155/2001/95404.e109a5ec4a8b47e8bc492fe12862f0c6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Prinz M, Heikenwalder M, Junt T, et al. Positioning of follicular dendritic cells within the spleen controls prion neuroinvasion. Nature. 2003;425:957–962. doi: 10.1038/nature02072. [DOI] [PubMed] [Google Scholar]
  • 61.Prinz M, Montrasio F, Klein MA, et al. Lymph nodal prion replication and neuroinvasion in mice devoid of follicular dendritic cells. Proc Natl Acad Sci U S A. 2002;99:919–924. doi: 10.1073/pnas.022626399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Montrasio F, Frigg R, Glatzel M, et al. Impaired prion replication in spleens of mice lacking functional follicular dendritic cells. Science. 2000;288:1257–1259. doi: 10.1126/science.288.5469.1257. [DOI] [PubMed] [Google Scholar]
  • 63.Bakkebo MK, Mouillet-Richard S, Espenes A, Goldmann W, Tatzelt J, Tranulis MA. The cellular prion protein: a player in immunological quiescence. Front Immunol. 2015;6:450. doi: 10.3389/fimmu.2015.00450.3dbe4c59310c4913aed7874866a17da0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Manni G, Lewis V, Senesi M, et al. The cellular prion protein beyond prion diseases. Swiss Med Wkly. 2020;150:w20222. doi: 10.4414/smw.2020.20222.99dd4a4ebbbf4db9b817115f13f4dfad [DOI] [PubMed] [Google Scholar]
  • 65.Ezpeleta J, Boudet-Devaud F, Pietri M, et al. Protective role of cellular prion protein against TNFalpha-mediated inflammation through TACE alpha-secretase. Sci Rep. 2017;7:7671. doi: 10.1038/s41598-017-08110-x.98b23f663c354ca58b1295e144d129b8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gadotti VM, Zamponi GW. Cellular prion protein protects from inflammatory and neuropathic pain. Mol Pain. 2011;7:59. doi: 10.1186/1744-8069-7-59.b1508c380fb6433a90a9dcc8dcb3f08a [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Shao J, Yin X, Lang Y, et al. Cellular prion protein attenuates OGD/R-induced damage by skewing microglia toward an anti-inflammatory state via enhanced and prolonged activation of autophagy. Mol Neurobiol. 2023;60:1297–1316. doi: 10.1007/s12035-022-03099-5. [DOI] [PubMed] [Google Scholar]
  • 68.Tsutsui S, Hahn JN, Johnson TA, Ali Z, Jirik FR. Absence of the cellular prion protein exacerbates and prolongs neuroinflammation in experimental autoimmune encephalomyelitis. Am J Pathol. 2008;173:1029–1041. doi: 10.2353/ajpath.2008.071062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Petit CS, Barreau F, Besnier L, et al. Requirement of cellular prion protein for intestinal barrier function and mislocalization in patients with inflammatory bowel disease. Gastroenterology. 2012;143:122–132. doi: 10.1053/j.gastro.2012.03.029. [DOI] [PubMed] [Google Scholar]
  • 70.Salvesen O, Reiten MR, Espenes A, Bakkebo MK, Tranulis MA, Ersdal C. LPS-induced systemic inflammation reveals an immunomodulatory role for the prion protein at the blood-brain interface. J Neuroinflammation. 2017;14:106. doi: 10.1186/s12974-017-0879-5.8a11d83250754409ae2642c0e409b8a6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liu J, Zhao D, Liu C, et al. Prion protein participates in the protection of mice from lipopolysaccharide infection by regulating the inflammatory process. J Mol Neurosci. 2015;55:279–287. doi: 10.1007/s12031-014-0319-2. [DOI] [PubMed] [Google Scholar]
  • 72.Pan Y, Zhao L, Liang J, et al. Cellular prion protein promotes invasion and metastasis of gastric cancer. FASEB J. 2006;20:1886–1888. doi: 10.1096/fj.06-6138fje. [DOI] [PubMed] [Google Scholar]
  • 73.Jiang B, Liu J, Lee MH. Targeting a designer TIMP-1 to the cell surface for effective mt1-mmp inhibition: a potential role for the prion protein in renal carcinoma therapy. Molecules. 2019;24:255. doi: 10.3390/molecules24020255.2d2dce1fb8b94e198ea67b15d2bf4e3b [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ong SH, Goh KW, Chieng CK, Say YH. Cellular prion protein and gamma-synuclein overexpression in LS 174T colorectal cancer cell drives endothelial proliferation-to-differentiation switch. PeerJ. 2018;6:e4506. doi: 10.7717/peerj.4506.57d720700ffb4da3bc2d3e8bf940d2c1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Antonacopoulou AG, Grivas PD, Skarlas L, Kalofonos M, Scopa CD, Kalofonos HP. POLR2F, ATP6V0A1 and PRNP expression in colorectal cancer: new molecules with prognostic significance? Anticancer Res. 2008;28:1221–1227. [PubMed] [Google Scholar]
  • 76.Roucou X, Giannopoulos PN, Zhang Y, Jodoin J, Goodyer CG, LeBlanc A. Cellular prion protein inhibits proapoptotic Bax conformational change in human neurons and in breast carcinoma MCF-7 cells. Cell Death Differ. 2005;12:783–795. doi: 10.1038/sj.cdd.4401629. [DOI] [PubMed] [Google Scholar]
  • 77.Meslin F, Hamai A, Gao P, et al. Silencing of prion protein sensitizes breast adriamycin-resistant carcinoma cells to TRAIL-mediated cell death. Cancer Res. 2007;67:10910–10919. doi: 10.1158/0008-5472.CAN-07-0512. [DOI] [PubMed] [Google Scholar]
  • 78.Cha S, Sin MJ, Kim MJ, et al. Involvement of cellular prion protein in invasion and metastasis of lung cancer by inducing treg cell development. Biomolecules. 2021;11:285. doi: 10.3390/biom11020285.db0eb7b92744473e889954183e7e5693 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Liang J, Pan Y, Zhang D, et al. Cellular prion protein promotes proliferation and G1/S transition of human gastric cancer cells SGC7901 and AGS. FASEB J. 2007;21:2247–2256. doi: 10.1096/fj.06-7799com. [DOI] [PubMed] [Google Scholar]
  • 80.Yap YH, Say YH. Resistance against tumour necrosis factor alpha apoptosis by the cellular prion protein is cell-specific for oral, colon and kidney cancer cell lines. Cell Biol Int. 2012;36:273–277. doi: 10.1042/CBI20110088. [DOI] [PubMed] [Google Scholar]
  • 81.Yun CW, Yun S, Lee JH, et al. Silencing prion protein in HT29 human colorectal cancer cells enhances anticancer response to fucoidan. Anticancer Res. 2016;36:4449–4458. doi: 10.21873/anticanres.10989. [DOI] [PubMed] [Google Scholar]
  • 82.Gil M, Kim YK, Kim KE, Kim W, Park CS, Lee KJ. Cellular prion protein regulates invasion and migration of breast cancer cells through MMP-9 activity. Biochem Biophys Res Commun. 2016;470:213–219. doi: 10.1016/j.bbrc.2016.01.038. [DOI] [PubMed] [Google Scholar]
  • 83.Boulay A, Masson R, Chenard MP, et al. High cancer cell death in syngeneic tumors developed in host mice deficient for the stromelysin-3 matrix metalloproteinase. Cancer Res. 2001;61:2189–2193. [PubMed] [Google Scholar]
  • 84.Wang Q, Qian J, Wang F, Ma Z. Cellular prion protein accelerates colorectal cancer metastasis via the Fyn-SP1-SATB1 axis. Oncol Rep. 2012;28:2029–2034. doi: 10.3892/or.2012.2025. [DOI] [PubMed] [Google Scholar]
  • 85.Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27:109–118. doi: 10.1038/cr.2016.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Shitara K, Nishikawa H. Regulatory T cells: a potential target in cancer immunotherapy. Ann N Y Acad Sci. 2018;1417:104–115. doi: 10.1111/nyas.13625. [DOI] [PubMed] [Google Scholar]
  • 87.Chen BJ, Zhao JW, Zhang DH, Zheng AH, Wu GQ. Immunotherapy of cancer by targeting regulatory T cells. Int Immunopharmacol. 2022;104:108469. doi: 10.1016/j.intimp.2021.108469. [DOI] [PubMed] [Google Scholar]

Articles from BMB Reports are provided here courtesy of Korean Society for Biochemistry and Molecular Biology

RESOURCES