1
|
Abi Nahed R, Safwan-Zaiter H, Gemy K, Lyko C, Boudaud M, Desseux M, Marquette C, Barjat T, Alfaidy N, Benharouga M. The Multifaceted Functions of Prion Protein (PrP C) in Cancer. Cancers (Basel) 2023; 15:4982. [PMID: 37894349 PMCID: PMC10605613 DOI: 10.3390/cancers15204982] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/23/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The cellular prion protein (PrPC) is a glycoprotein anchored to the cell surface by glycosylphosphatidylinositol (GPI). PrPC is expressed both in the brain and in peripheral tissues. Investigations on PrPC's functions revealed its direct involvement in neurodegenerative and prion diseases, as well as in various physiological processes such as anti-oxidative functions, copper homeostasis, trans-membrane signaling, and cell adhesion. Recent findings have revealed the ectopic expression of PrPC in various cancers including gastric, melanoma, breast, colorectal, pancreatic, as well as rare cancers, where PrPC promotes cellular migration and invasion, tumor growth, and metastasis. Through its downstream signaling, PrPC has also been reported to be involved in resistance to chemotherapy and tumor cell apoptosis. This review summarizes the variance of expression of PrPC in different types of cancers and discusses its roles in their development and progression, as well as its use as a potential target to treat such cancers.
Collapse
Affiliation(s)
- Roland Abi Nahed
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Hasan Safwan-Zaiter
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Kevin Gemy
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Camille Lyko
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Mélanie Boudaud
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Morgane Desseux
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Christel Marquette
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Tiphaine Barjat
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Nadia Alfaidy
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| | - Mohamed Benharouga
- U1292, Laboratoire de BioSanté, Institut National de la Santé et de la Recherche Médicale (INSERM), F-38058 Grenoble, France; (R.A.N.); (H.S.-Z.); (K.G.); (C.L.); (M.B.); (M.D.); (C.M.); (T.B.); (N.A.)
- Commissariat à l’Energie Atomique (CEA), DSV-IRIG, F-38054 Grenoble, France
- University of Grenoble Alpes (UGA), F-38058 Grenoble, France
| |
Collapse
|
2
|
Yousaf S, Ahmad M, Wu S, Zia MA, Ahmed I, Iqbal HMN, Liu Q, Rehman SU. Cellular Prion Protein Role in Cancer Biology: Is It A Potential Therapeutic Target? Biomedicines 2022; 10:2833. [PMID: 36359353 PMCID: PMC9687521 DOI: 10.3390/biomedicines10112833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 11/09/2022] Open
Abstract
Cancers are worldwide health concerns, whether they are sporadic or hereditary. The fundamental mechanism that causes somatic or oncogenic mutations and ultimately aids cancer development is still unknown. However, mammalian cells with protein-only somatic inheritance may also contribute to cancerous malignancies. Emerging data from a recent study show that prion-like proteins and prions (PrPC) are crucial entities that have a functional role in developing neurological disorders and cancer. Furthermore, excessive PrPC expression profiling has also been detected in non-neuronal tissues, such as the lymphoid cells, kidney, GIT, lung, muscle, and mammary glands. PrPC expression is strongly linked with the proliferation and metastasis of pancreatic, prostate, colorectal, and breast malignancies. Similarly, experimental investigation presented that the PrPC expression, including the prion protein-coding gene (PRNP) and p53 ag are directly associated with tumorigenicity and metastasis (tumor suppressor gene). The ERK2 (extracellular signal-regulated kinase) pathway also confers a robust metastatic capability for PrPC-induced epithelial to mesenchymal transition. Additionally, prions could alter the epigenetic regulation of genes and overactive the mitogen-activated protein kinase (MAPK) signaling pathway, which promotes the development of cancer in humans. Protein overexpression or suppression caused by a prion and prion-like proteins has also been linked to oncogenesis and metastasis. Meanwhile, additional studies have discovered resistance to therapeutic targets, highlighting the significance of protein expression levels as potential diagnostic indicators and therapeutic targets.
Collapse
Affiliation(s)
- Saba Yousaf
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources of Guangxi University, Nanning 530005, China
- Enzyme Biotechnology Laboratory, Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad 38040, Pakistan
| | - Muhammad Ahmad
- Faculty of Veterinary Sciences, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences (SBBUVAS), Sakrand 67210, Pakistan
| | - Siwen Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources of Guangxi University, Nanning 530005, China
| | - Muhammad Anjum Zia
- Enzyme Biotechnology Laboratory, Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad 38040, Pakistan
| | - Ishtiaq Ahmed
- Department of Regional Science Operations, La Trobe Rural Health School, Albury-Wodonga, VIC 3689, Australia
| | - Hafiz M. N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources of Guangxi University, Nanning 530005, China
| | - Saif ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources of Guangxi University, Nanning 530005, China
| |
Collapse
|
3
|
The Cellular Prion Protein: A Promising Therapeutic Target for Cancer. Int J Mol Sci 2020; 21:ijms21239208. [PMID: 33276687 PMCID: PMC7730109 DOI: 10.3390/ijms21239208] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/28/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022] Open
Abstract
Studies on the cellular prion protein (PrPC) have been actively conducted because misfolded PrPC is known to cause transmissible spongiform encephalopathies or prion disease. PrPC is a glycophosphatidylinositol-anchored cell surface glycoprotein that has been reported to affect several cellular functions such as stress protection, cellular differentiation, mitochondrial homeostasis, circadian rhythm, myelin homeostasis, and immune modulation. Recently, it has also been reported that PrPC mediates tumor progression by enhancing the proliferation, metastasis, and drug resistance of cancer cells. In addition, PrPC regulates cancer stem cell properties by interacting with cancer stem cell marker proteins. In this review, we summarize how PrPC promotes tumor progression in terms of proliferation, metastasis, drug resistance, and cancer stem cell properties. In addition, we discuss strategies to treat tumors by modulating the function and expression of PrPC via the regulation of HSPA1L/HIF-1α expression and using an anti-prion antibody.
Collapse
|
4
|
Physiological role of Prion Protein in Copper homeostasis and angiogenic mechanisms of endothelial cells. THE EUROBIOTECH JOURNAL 2019. [DOI: 10.2478/ebtj-2019-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
The Prion Protein (PrP) is mostly known for its role in prion diseases, where its misfolding and aggregation can cause fatal neurodegenerative conditions such as the bovine spongiform encephalopathy and human Creutzfeldt–Jakob disease. Physiologically, PrP is involved in several processes including adhesion, proliferation, differentiation and angiogenesis, but the molecular mechanisms behind its role remain unclear. PrP, due to its well-described structure, is known to be able to regulate copper homeostasis; however, copper dyshomeostasis can lead to developmental defects. We investigated PrP-dependent regulation of copper homeostasis in human endothelial cells (HUVEC) using an RNA-interference protocol. PrP knockdown did not influence cell viability in silenced HUVEC (PrPKD) compared to control cells, but significantly increased PrPKD HUVEC cells sensitivity to cytotoxic copper concentrations. A reduction of PrPKD cells reductase activity and copper ions transport capacity was observed. Furthermore, PrPKD-derived spheroids exhibited altered morphogenesis and their derived cells showed a decreased vitality 24 and 48 hours after seeding. PrPKD spheroid-derived cells also showed disrupted tubulogenesis in terms of decreased coverage area, tubule length and total nodes number on matrigel, preserving unaltered VEGF receptors expression levels. Our results highlight PrP physiological role in cellular copper homeostasis and in the angiogenesis of endothelial cells.
Collapse
|
5
|
The function of the cellular prion protein in health and disease. Acta Neuropathol 2018; 135:159-178. [PMID: 29151170 DOI: 10.1007/s00401-017-1790-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022]
Abstract
The essential role of the cellular prion protein (PrPC) in prion disorders such as Creutzfeldt-Jakob disease is well documented. Moreover, evidence is accumulating that PrPC may act as a receptor for protein aggregates and transduce neurotoxic signals in more common neurodegenerative disorders, such as Alzheimer's disease. Although the pathological roles of PrPC have been thoroughly characterized, a general consensus on its physiological function within the brain has not yet been established. Knockout studies in various organisms, ranging from zebrafish to mice, have implicated PrPC in a diverse range of nervous system-related activities that include a key role in the maintenance of peripheral nerve myelination as well as a general ability to protect against neurotoxic stimuli. Thus, the function of PrPC may be multifaceted, with different cell types taking advantage of unique aspects of its biology. Deciphering the cellular function(s) of PrPC and the consequences of its absence is not simply an academic curiosity, since lowering PrPC levels in the brain is predicted to be a powerful therapeutic strategy for the treatment of prion disease. In this review, we outline the various approaches that have been employed in an effort to uncover the physiological and pathological functions of PrPC. While these studies have revealed important clues about the biology of the prion protein, the precise reason for PrPC's existence remains enigmatic.
Collapse
|
6
|
Reiten MR, Malachin G, Kommisrud E, Østby GC, Waterhouse KE, Krogenæs AK, Kusnierczyk A, Bjørås M, Jalland CMO, Nekså LH, Røed SS, Stenseth EB, Myromslien FD, Zeremichael TT, Bakkebø MK, Espenes A, Tranulis MA. Stress Resilience of Spermatozoa and Blood Mononuclear Cells without Prion Protein. Front Mol Biosci 2018; 5:1. [PMID: 29417049 PMCID: PMC5787566 DOI: 10.3389/fmolb.2018.00001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/08/2018] [Indexed: 11/19/2022] Open
Abstract
The cellular prion protein PrPC is highly expressed in neurons, but also present in non-neuronal tissues, including the testicles and spermatozoa. Most immune cells and their bone marrow precursors also express PrPC. Clearly, this protein operates in highly diverse cellular contexts. Investigations into putative stress-protective roles for PrPC have resulted in an array of functions, such as inhibition of apoptosis, stimulation of anti-oxidant enzymes, scavenging roles, and a role in nuclear DNA repair. We have studied stress resilience of spermatozoa and peripheral blood mononuclear cells (PBMCs) derived from non-transgenic goats that lack PrPC (PRNPTer/Ter) compared with cells from normal (PRNP+/+) goats. Spermatozoa were analyzed for freeze tolerance, DNA integrity, viability, motility, ATP levels, and acrosome intactness at rest and after acute stress, induced by Cu2+ ions, as well as levels of reactive oxygen species (ROS) after exposure to FeSO4 and H2O2. Surprisingly, PrPC-negative spermatozoa reacted similarly to normal spermatozoa in all read-outs. Moreover, in vitro exposure of PBMCs to Doxorubicin, H2O2 and methyl methanesulfonate (MMS), revealed no effect of PrPC on cellular survival or global accumulation of DNA damage. Similar results were obtained with human neuroblastoma (SH-SY5Y) cell lines stably expressing varying levels of PrPC. RNA sequencing of PBMCs (n = 8 of PRNP+/+ and PRNPTer/Ter) showed that basal level expression of genes encoding DNA repair enzymes, ROS scavenging, and antioxidant enzymes were unaffected by the absence of PrPC. Data presented here questions the in vitro cytoprotective roles previously attributed to PrPC, although not excluding such functions in other cell types or tissues during inflammatory stress.
Collapse
Affiliation(s)
- Malin R Reiten
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Giulia Malachin
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Elisabeth Kommisrud
- Faculty of Education and Natural Sciences, Inland University of Applied Sciences, Hamar, Norway
| | - Gunn C Østby
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Karin E Waterhouse
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway.,Spermvital AS Holsetgata, Hamar, Norway
| | - Anette K Krogenæs
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Anna Kusnierczyk
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Clara M O Jalland
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Liv Heidi Nekså
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Susan S Røed
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Else-Berit Stenseth
- Faculty of Education and Natural Sciences, Inland University of Applied Sciences, Hamar, Norway
| | - Frøydis D Myromslien
- Faculty of Education and Natural Sciences, Inland University of Applied Sciences, Hamar, Norway
| | - Teklu T Zeremichael
- Faculty of Education and Natural Sciences, Inland University of Applied Sciences, Hamar, Norway
| | - Maren K Bakkebø
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Arild Espenes
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Michael A Tranulis
- Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| |
Collapse
|
7
|
Ji Z, Wang B, Yan K, Dong L, Meng G, Shi L. A linear programming computational framework integrates phosphor-proteomics and prior knowledge to predict drug efficacy. BMC SYSTEMS BIOLOGY 2017; 11:127. [PMID: 29322918 PMCID: PMC5763468 DOI: 10.1186/s12918-017-0501-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background In recent years, the integration of ‘omics’ technologies, high performance computation, and mathematical modeling of biological processes marks that the systems biology has started to fundamentally impact the way of approaching drug discovery. The LINCS public data warehouse provides detailed information about cell responses with various genetic and environmental stressors. It can be greatly helpful in developing new drugs and therapeutics, as well as improving the situations of lacking effective drugs, drug resistance and relapse in cancer therapies, etc. Results In this study, we developed a Ternary status based Integer Linear Programming (TILP) method to infer cell-specific signaling pathway network and predict compounds’ treatment efficacy. The novelty of our study is that phosphor-proteomic data and prior knowledge are combined for modeling and optimizing the signaling network. To test the power of our approach, a generic pathway network was constructed for a human breast cancer cell line MCF7; and the TILP model was used to infer MCF7-specific pathways with a set of phosphor-proteomic data collected from ten representative small molecule chemical compounds (most of them were studied in breast cancer treatment). Cross-validation indicated that the MCF7-specific pathway network inferred by TILP were reliable predicting a compound’s efficacy. Finally, we applied TILP to re-optimize the inferred cell-specific pathways and predict the outcomes of five small compounds (carmustine, doxorubicin, GW-8510, daunorubicin, and verapamil), which were rarely used in clinic for breast cancer. In the simulation, the proposed approach facilitates us to identify a compound’s treatment efficacy qualitatively and quantitatively, and the cross validation analysis indicated good accuracy in predicting effects of five compounds. Conclusions In summary, the TILP model is useful for discovering new drugs for clinic use, and also elucidating the potential mechanisms of a compound to targets. Electronic supplementary material The online version of this article (10.1186/s12918-017-0501-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhiwei Ji
- School of Electronical and Information Engineering, Anhui University of Technology, Maanshan, 243002, China. .,School of Information & Electronic Engineering, Zhejiang Gongshang University, 18 Xuezheng Road, Hangzhou, 310018, China.
| | - Bing Wang
- School of Electronical and Information Engineering, Anhui University of Technology, Maanshan, 243002, China.
| | - Ke Yan
- College of Information Engineering, China Jiliang University, 258 Xueyuan Streat, Hangzhou, 310018, China
| | - Ligang Dong
- School of Information & Electronic Engineering, Zhejiang Gongshang University, 18 Xuezheng Road, Hangzhou, 310018, China
| | - Guanmin Meng
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, 234 Gucui Road, Hangzhou, 310012, China
| | - Lei Shi
- School of Information & Electronic Engineering, Zhejiang Gongshang University, 18 Xuezheng Road, Hangzhou, 310018, China
| |
Collapse
|
8
|
Castle AR, Gill AC. Physiological Functions of the Cellular Prion Protein. Front Mol Biosci 2017; 4:19. [PMID: 28428956 PMCID: PMC5382174 DOI: 10.3389/fmolb.2017.00019] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/22/2017] [Indexed: 01/09/2023] Open
Abstract
The prion protein, PrPC, is a small, cell-surface glycoprotein notable primarily for its critical role in pathogenesis of the neurodegenerative disorders known as prion diseases. A hallmark of prion diseases is the conversion of PrPC into an abnormally folded isoform, which provides a template for further pathogenic conversion of PrPC, allowing disease to spread from cell to cell and, in some circumstances, to transfer to a new host. In addition to the putative neurotoxicity caused by the misfolded form(s), loss of normal PrPC function could be an integral part of the neurodegenerative processes and, consequently, significant research efforts have been directed toward determining the physiological functions of PrPC. In this review, we first summarise important aspects of the biochemistry of PrPC before moving on to address the current understanding of the various proposed functions of the protein, including details of the underlying molecular mechanisms potentially involved in these functions. Over years of study, PrPC has been associated with a wide array of different cellular processes and many interacting partners have been suggested. However, recent studies have cast doubt on the previously well-established links between PrPC and processes such as stress-protection, copper homeostasis and neuronal excitability. Instead, the functions best-supported by the current literature include regulation of myelin maintenance and of processes linked to cellular differentiation, including proliferation, adhesion, and control of cell morphology. Intriguing connections have also been made between PrPC and the modulation of circadian rhythm, glucose homeostasis, immune function and cellular iron uptake, all of which warrant further investigation.
Collapse
|
9
|
Tang Z, Ma J, Zhang W, Gong C, He J, Wang Y, Yu G, Yuan C, Wang X, Sun Y, Ma J, Liu F, Zhao Y. The Role of Prion Protein Expression in Predicting Gastric Cancer Prognosis. J Cancer 2016; 7:984-90. [PMID: 27313789 PMCID: PMC4910591 DOI: 10.7150/jca.14237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/15/2016] [Indexed: 12/26/2022] Open
Abstract
Previous reports indicated that prion protein (PrP) is involved in gastric cancer (GC) development and progression, but its role in GC prognosis has been poorly characterized. A total of 480 GC patients were recruited in this retrospective study. PrP expression in cancerous and non-cancerous gastric tissues was detected by using the tissue microarray and immunohistochemical staining techniques. Our results showed that the PrP expression in GC was significantly less frequent than that in the non-cancerous gastric tissue (44.4% vs 66.4%, P < 0.001). Cox regression analysis revealed that PrP expression was associated with TNM stage, survival status and survival time. GC patients with higher TNM stages (stages II, III and IV) had significantly lower PrP expression levels in tumors than those with lower TNM stages (stages 0 and I). Kaplan-Meier survival curves revealed that negative PrP expression was associated with poor overall survival (log-rank test: P < 0.001). The mean survival time for patients with negative PrP expression was significant lower than those with positive PrP expression (43.0±28.5m vs. 53.9±31.1m, P<0.001). In multivariate Cox hazard regression, PrP expression was an independent prognostic factor for GC survival, with a HR (hazard ratio) of 0.687 (95%CI:0.520-0.907, P=0.008). Our results revealed that negative PrP expression could independently predict worse outcome in GC and thereby could be used to guide the clinical practice.
Collapse
Affiliation(s)
- Zhaoqing Tang
- 1. Department of General surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ji Ma
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Wei Zhang
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China;; 5. Department of Pathology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, P.R. China
| | - Changguo Gong
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Jing He
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Ying Wang
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China;; 3. Department of Physiology, Renji College, Wenzhou Medical University, Wenzhou, P.R. China
| | - Guohua Yu
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Chonggang Yuan
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Xuefei Wang
- 1. Department of General surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yihong Sun
- 1. Department of General surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jiyan Ma
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China;; 4. Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Fenglin Liu
- 1. Department of General surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yulan Zhao
- 2. School of Life Sciences, East China Normal University, Shanghai, P.R. China
| |
Collapse
|
10
|
Cellular prion protein contributes to LS 174T colon cancer cell carcinogenesis by increasing invasiveness and resistance against doxorubicin-induced apoptosis. Tumour Biol 2015; 36:8107-20. [PMID: 25983001 DOI: 10.1007/s13277-015-3530-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/04/2015] [Indexed: 12/17/2022] Open
Abstract
As the cellular prion protein (PrP(C)) has been implicated in carcinogenesis, we aimed to investigate the effects of cancer cell-specific PrP(C) overexpression from the invasion, metastasis, and apoptosis aspects, by performing cell motility assays, cell proliferation assays under anchorage-dependent and anchorage-independent conditions, and apoptosis evasion when subjected to multiple anti-cancer drugs. Overexpression of PrP(C) in LS 174T was achieved by stable transfection. PrP(C) overexpression was shown to increase cell proliferation in anchorage-dependent and anchorage-independent manners, as shown by more viable cells in 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, more colonies formed in soft agar assay and increased resistance to anoikis in poly-2-hydroxyethyl methacrylate-coated surface. PrP(C) overexpression also increased cell motility and invasiveness of LS 174T. Cell adhesion to extracellular matrix using collagen- and fibronectin-coated surfaces revealed increased cell attachment in LS 174T cells overexpressing PrP(C). Analysis of apoptotic and necrotic cells by propidium iodide/annexin V-fluorescein isothiocyanate microscopy and 7-amino-actinomycin D/annexin V-phycoerythrin flow cytometry revealed that PrP(C) overexpression attenuated doxorubicin-induced apoptosis. Human apoptosis antibody array with 35 apoptosis-related proteins revealed that three inhibitor of apoptosis proteins (IAPs)-survivin, X-linked inhibitor of apoptosis protein (XIAP), and cellular inhibitor of apoptosis protein-1 (cIAP-1)-were upregulated in LS 174T cells overexpressing PrP(C) in doxorubicin-induced apoptosis. In conclusion, the overexpression of PrP(C) could enhance the invasiveness and survival of LS 174T colorectal cancer cells, indicating that PrP(C) plays a role in colorectal cancer biology.
Collapse
|
11
|
Disruption of prion protein-HOP engagement impairs glioblastoma growth and cognitive decline and improves overall survival. Oncogene 2014; 34:3305-14. [PMID: 25151961 DOI: 10.1038/onc.2014.261] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 07/03/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022]
Abstract
Glioblastomas (GBMs) are resistant to current therapy protocols and identification of molecules that target these tumors is crucial. Interaction of secreted heat-shock protein 70 (Hsp70)-Hsp90-organizing protein (HOP) with cellular prion protein (PrP(C)) triggers a large number of trophic effects in the nervous system. We found that both PrP(C) and HOP are highly expressed in human GBM samples relative to non-tumoral tissue or astrocytoma grades I-III. High levels of PrP(C) and HOP were associated with greater GBM proliferation and lower patient survival. HOP-PrP(C) binding increased GBM proliferation in vitro via phosphatidylinositide 3-kinase and extracellular-signal-regulated kinase pathways, and a HOP peptide mimicking the PrP(C) binding site (HOP230-245) abrogates this effect. PrP(C) knockdown impaired tumor growth and increased survival of mice with tumors. In mice, intratumor delivery of HOP230-245 peptide impaired proliferation and promoted apoptosis of GBM cells. In addition, treatment with HOP230-245 peptide inhibited tumor growth, maintained cognitive performance and improved survival. Thus, together, the present results indicate that interfering with PrP(C)-HOP engagement is a promising approach for GBM therapy.
Collapse
|
12
|
Yang X, Zhang Y, Zhang L, He T, Zhang J, Li C. Prion protein and cancers. Acta Biochim Biophys Sin (Shanghai) 2014; 46:431-40. [PMID: 24681883 DOI: 10.1093/abbs/gmu019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The normal cellular prion protein, PrP(C) is a highly conserved and widely expressed cell surface glycoprotein in all mammals. The expression of PrP is pivotal in the pathogenesis of prion diseases; however, the normal physiological functions of PrP(C) remain incompletely understood. Based on the studies in cell models, a plethora of functions have been attributed to PrP(C). In this paper, we reviewed the potential roles that PrP(C) plays in cell physiology and focused on its contribution to tumorigenesis.
Collapse
Affiliation(s)
- Xiaowen Yang
- Department of the First Abdominal Surgery, Jiangxi Tumor Hospital, Nanchang 330029, China
| | - Yan Zhang
- Department of Molecular Endocrinology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Lihua Zhang
- Department of Pathology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Tianlin He
- Department of General Surgery, Changhai Hospital of Second Military Medical University, Shanghai 200433, China
| | - Jie Zhang
- Department of Stomatology, The First Affiliated Hospital of Shihezi University Medical College, Shihezi 832000, China
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|