1
|
Cheon YP, Ryou C, Svedružić ŽM. Roles of prion proteins in mammalian development. Anim Cells Syst (Seoul) 2024; 28:551-566. [PMID: 39664939 PMCID: PMC11633422 DOI: 10.1080/19768354.2024.2436860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/02/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
Prion protein (PrP) is highly conserved and is expressed in most tissues in a developmental stage-specific manner. Glycosylated cellular prion protein (PrPC) is found in most cells and subcellular areas as a physiological regulating molecule. On the other hand, the amyloid form of PrPC, scrapie PrP (PrPSC), causes transmissible pathogenesis in the central nervous system and induces degeneration of the nervous system. Although many amyloids are reversible and critical in determining the fate, differentiation, and physiological functions of cells, thus far, PrPSC originating from PrPC is not. Although many studies have focused on disorders involving PrPC and the deletion mammalian models for PrPC have no severe phenotype, it has been suggested that PrPC has a role in normal development. It is conserved and expressed from gametes to adult somatic cells. In addition, severe developmental phenotypes appear in PrP null zebrafish embryos and in various mammalian cell model systems. In addition, it has been well established that PrPC is strongly involved in the stemness and differentiation of embryonic stem cells and progenitors. Thus far, many studies on PrPC have focused mostly on disease-associated conditions with physiological roles as a complex platform but not on development. The known roles of PrPC depend on the interacting molecules through its flexible tail and domains. PrPC interacts with membrane, and various intracellular and extracellular molecules. In addition, PrPC and amyloid can stimulate signaling pathways differentially. In this review, we summarize the function of prion protein and discuss its role in development.
Collapse
Affiliation(s)
- Yong-Pil Cheon
- Division of Developmental Biology and Physiology, Department of Biotechnology, Institute for Basic Sciences, Sungshin University, Seoul, Korea
| | - Chongsuk Ryou
- Department of Pharmacy, College of Pharmacy, Hanyang University, ekcho Ansan, Korea
| | | |
Collapse
|
2
|
Groveman BR, Schwarz B, Bohrnsen E, Foliaki ST, Carroll JA, Wood AR, Bosio CM, Haigh CL. A PrP EGFR signaling axis controls neural stem cell senescence through modulating cellular energy pathways. J Biol Chem 2023; 299:105319. [PMID: 37802314 PMCID: PMC10641666 DOI: 10.1016/j.jbc.2023.105319] [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: 04/21/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/08/2023] Open
Abstract
Mis-folding of the prion protein (PrP) is known to cause neurodegenerative disease; however, the native function of this protein remains poorly defined. PrP has been linked with many cellular functions, including cellular proliferation and senescence. It is also known to influence epidermal growth factor receptor (EGFR) signaling, a pathway that is itself linked with both cell growth and senescence. Adult neural stem cells (NSCs) persist at low levels in the brain throughout life and retain the ability to proliferate and differentiate into new neural lineage cells. KO of PrP has previously been shown to reduce NSC proliferative capacity. We used PrP KO and WT NSCs from adult mouse brain to examine the influence of PrP on cellular senescence, EGFR signaling, and the downstream cellular processes. PrP KO NSCs showed decreased cell proliferation and increased senescence in in vitro cultures. Expression of EGFR was decreased in PrP KO NSCs compared with WT NSCs and additional supplementation of EGF was sufficient to reduce senescence. RNA-seq analysis confirmed that significant changes were occurring at the mRNA level within the EGFR signaling pathway and these were associated with reduced expression of mitochondrial components and correspondingly reduced mitochondrial function. Metabolomic analysis of cellular energy pathways showed that blockages were occurring at critical sites for production of energy and biomass, including catabolism of pyruvate. We conclude that, in the absence of PrP, NSC growth pathways are downregulated as a consequence of insufficient energy and growth intermediates.
Collapse
Affiliation(s)
- Bradley R Groveman
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Benjamin Schwarz
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Eric Bohrnsen
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Simote T Foliaki
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - James A Carroll
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Aleksandar R Wood
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Catharine M Bosio
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Cathryn L Haigh
- Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA.
| |
Collapse
|
3
|
Bruce MR, Couch ACM, Grant S, McLellan J, Ku K, Chang C, Bachman A, Matson M, Berman RF, Maddock RJ, Rowland D, Kim E, Ponzini MD, Harvey D, Taylor SL, Vernon AC, Bauman MD, Van de Water J. Altered behavior, brain structure, and neurometabolites in a rat model of autism-specific maternal autoantibody exposure. Mol Psychiatry 2023; 28:2136-2147. [PMID: 36973347 PMCID: PMC10575787 DOI: 10.1038/s41380-023-02020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023]
Abstract
Maternal immune dysregulation is a prenatal risk factor for autism spectrum disorder (ASD). Importantly, a clinically relevant connection exists between inflammation and metabolic stress that can result in aberrant cytokine signaling and autoimmunity. In this study we examined the potential for maternal autoantibodies (aAbs) to disrupt metabolic signaling and induce neuroanatomical changes in the brains of exposed offspring. To accomplish this, we developed a model of maternal aAb exposure in rats based on the clinical phenomenon of maternal autoantibody-related ASD (MAR-ASD). Following confirmation of aAb production in rat dams and antigen-specific immunoglobulin G (IgG) transfer to offspring, we assessed offspring behavior and brain structure longitudinally. MAR-ASD rat offspring displayed a reduction in pup ultrasonic vocalizations and a pronounced deficit in social play behavior when allowed to freely interact with a novel partner. Additionally, longitudinal in vivo structural magnetic resonance imaging (sMRI) at postnatal day 30 (PND30) and PND70, conducted in a separate cohort of animals, revealed sex-specific differences in total and regional brain volume. Treatment-specific effects by region appeared to converge on midbrain and cerebellar structures in MAR-ASD offspring. Simultaneously, in vivo 1H magnetic resonance spectroscopy (1H-MRS) data were collected to examine brain metabolite levels in the medial prefrontal cortex. Results showed that MAR-ASD offspring displayed decreased levels of choline-containing compounds and glutathione, accompanied by increased taurine compared to control animals. Overall, we found that rats exposed to MAR-ASD aAbs present with alterations in behavior, brain structure, and neurometabolites; reminiscent of findings observed in clinical ASD.
Collapse
Affiliation(s)
- Matthew R Bruce
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Amalie C M Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Simone Grant
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Janna McLellan
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA
| | - Katherine Ku
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Christina Chang
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Angelica Bachman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Matthew Matson
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Robert F Berman
- Department of Neurological Surgery, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Douglas Rowland
- Center for Molecular and Genomic Imaging, University of California, Davis, CA, USA
| | - Eugene Kim
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Matthew D Ponzini
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Danielle Harvey
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Sandra L Taylor
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
| | - Judy Van de Water
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Clinical Immunology, University of California, Davis, CA, USA.
- MIND Institute, University of California, Davis, CA, USA.
| |
Collapse
|
4
|
Abstract
The Hsp70/Hsp90 organising protein (Hop, also known as stress-inducible protein 1/STI1/STIP1) has received considerable attention for diverse cellular functions in both healthy and diseased states. There is extensive evidence that intracellular Hop is a co-chaperone of the major chaperones Hsp70 and Hsp90, playing an important role in the productive folding of Hsp90 client proteins, although recent evidence suggests that eukaryotic Hop is regulatory within chaperone complexes rather than essential. Consequently, Hop is implicated in many key signalling pathways, including aberrant pathways leading to cancer. Hop is also secreted, and it is now well established that Hop interacts with the prion protein, PrPC, to mediate multiple signalling events. The intracellular and extracellular forms of Hop most likely represent two different isoforms, although the molecular determinants of these divergent functions are yet to be identified. There is also a growing body of research that reports the involvement of Hop in cellular activities that appear independent of either chaperones or PrPC. While the various cellular functions of Hop have been described, its biological function remains elusive. However, recent knockout studies in mammals suggest that Hop has an important role in embryonic development. This review provides a critical overview of the latest molecular, cellular and biological research on Hop, critically evaluating its function in healthy systems and how this function is adapted in diseased states.
Collapse
|
5
|
Kim YC, Jeong BH. Transcriptomic analysis identifies novel potential biomarkers and highlights cilium-related biological processes in the early stages of prion disease in mice. Prion 2022; 16:84-90. [PMID: 35786398 PMCID: PMC9255203 DOI: 10.1080/19336896.2022.2095186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Prion diseases are fatal and irreversible neurodegenerative diseases induced by the pathogenic form of the prion protein (PrPSc), which is converted from the benign form of the prion protein (PrPC). These diseases are characterized by an extended asymptomatic incubation period accompanied by continuous conversion of PrPC to PrPSc. However, to date, the mechanism governing the conversion to PrPSc in the initial stages of prion disease has not been fully elucidated. We collected transcriptome data from the hippocampus of wild-type mice and prion-infected mice at 8 weeks post injection from the Gene Expression Omnibus and analysed differentially expressed genes and related signalling biological process using bioinformatic tools. We identified a total of 36 differentially expressed genes, including 22 upregulated genes and 14 downregulated genes. In addition, we identified that the cilium-related biological process was enriched in the early stages of prion disease. Furthermore, up- and down-regulated genes were associated with cilium-related cellular components and synapse-related cellular components, respectively. To the best of our knowledge, our study was the first to observe the upregulation of cilium-related genes in the early stages of prion disease.
Collapse
Affiliation(s)
- Yong-Chan Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Republic of Korea,Department of Bioactive Material Sciences and Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju, Republic of Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Republic of Korea,Department of Bioactive Material Sciences and Institute for Molecular Biology and Genetics, Jeonbuk National University, Jeonju, Republic of Korea,CONTACT Byung-Hoon Jeong Korea Zoonosis Research Institute, Jeonbuk National University, 820-120, Hana-ro, Iksan, Jeonbuk54531, Republic of Korea
| |
Collapse
|
6
|
Wu X, Liu M, Yan T, Wang Z, Yu W, Du Q, Hu W, Zheng Y, Zhang Z, Wang K, Dong X. Plasma PRPC Levels Correlate With Severity and Prognosis of Intracerebral Hemorrhage. Front Neurol 2022; 13:913926. [PMID: 35899267 PMCID: PMC9309369 DOI: 10.3389/fneur.2022.913926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundCellular prion protein (PRPC) exerts brain-protective effects. We determined the relationship between plasma PRPC levels and disease severity plus clinical outcome after acute intracerebral hemorrhage (ICH).MethodsA total of 138 ICH patients and 138 healthy controls were included in this prospective, observational study. Hematoma volume and Glasgow coma scale (GCS) score were used to assess disease severity. Glasgow outcome scale (GOS) scores of 1–3 and 4–5 at 90 days after stroke were defined as a poor outcome and good outcome, respectively. Using multivariate analysis, we discerned the relation of plasma PRPC levels to disease severity and poor outcome. The receiver operating characteristic (ROC) curve was built to evaluate the prognostic predictive capability.ResultsPlasma PRPC levels in ICH patients were significantly higher than those in healthy controls (median, 4.20 vs. 2.02 ng/ml; P < 0.001), and were independently correlated with GCS score (r = −0.645, P < 0.001) and hematoma volume (r = 0.627, P < 0.001). Plasma PRPC levels were highly correlated with GOS score (r = −0.762, P < 0.001), and were substantially higher in patients with poor outcomes than in those with the good outcomes. Using maximum Youden index, plasma PRPC levels >3.893 ng/ml distinguished the risk of poor outcome at 90 days, with a sensitivity of 86.4% and a specificity of 65.8% (area under the curve, 0.809; 95% confidence interval (CI), 0.737–0.881, P < 0.001). Plasma PRPC levels >3.893 ng/ml were independently associated with a poor 90-day outcome with an odds ratio of 12.278 (95% CI, 5.101–29.554).ConclusionElevated plasma PRPC levels are significantly associated with disease severity and poor 90-day outcome in ICH patients, indicating that plasma PRPC may be used as a potential prognostic biomarker after ICH.
Collapse
Affiliation(s)
- Xiaoyu Wu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ming Liu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tian Yan
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zefan Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenhua Yu
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Quan Du
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Hu
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongke Zheng
- Department of Intensive Care Unit, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zuyong Zhang
- Department of Neurosurgery, Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Keyi Wang
- Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Keyi Wang
| | - Xiaoqiao Dong
- Department of Neurosurgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Xiaoqiao Dong
| |
Collapse
|
7
|
Bianchini M, Giambelluca MA, Scavuzzo MC, Di Franco G, Guadagni S, Palmeri M, Furbetta N, Gianardi D, Funel N, Ricci C, Gaeta R, Pollina LE, Falcone A, Vivaldi C, Di Candio G, Biagioni F, Busceti CL, Morelli L, Fornai F. Detailing the ultrastructure's increase of prion protein in pancreatic adenocarcinoma. World J Gastroenterol 2021; 27:7324-7339. [PMID: 34876792 PMCID: PMC8611201 DOI: 10.3748/wjg.v27.i42.7324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/14/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Recent evidences have shown a relationship between prion protein (PrPc) expression and pancreatic ductal adenocarcinoma (PDAC). Indeed, PrPc could be one of the markers explaining the aggressiveness of this tumor. However, studies investigating the specific compartmentalization of increased PrPc expression within PDAC cells are lacking, as well as a correlation between ultrastructural evidence, ultrastructural morphometry of PrPc protein and clinical data. These data, as well as the quantitative stoichiometry of this protein detected by immuno-gold, provide a significant advancement in understanding the biology of disease and the outcome of surgical resection. AIM To analyze quantitative stoichiometry and compartmentalization of PrPc in PDAC cells and to correlate its presence with prognostic data. METHODS Between June 2018 and December 2020, samples from pancreatic tissues of 45 patients treated with pancreatic resection for a preoperative suspicion of PDAC at our Institution were collected. When the frozen section excluded a PDAC diagnosis, or the nodules were too small for adequate sampling, patients were ruled out from the present study. Western blotting was used to detect, quantify and compare the expression of PrPc in PDAC and control tissues, such as those of non-affected neighboring pancreatic tissue of the same patient. To quantify the increase of PrPc and to detect the subcellular compartmentalization of PrPc within PDAC cells, immuno-gold stoichiometry within specific cell compartments was analyzed with electron microscopy. Finally, an analysis of quantitative PrPc expression according to prognostic data, such as cancer stage, recurrence of the disease at 12 mo after surgery and recurrence during adjuvant chemotherapy was made. RESULTS The amount of PrPc within specimen from 38 out of 45 patients was determined by semi-quantitative analysis by using Western blotting, which indicates that PrPc increases almost three-fold in tumor pancreatic tissue compared with healthy pancreatic regions [242.41 ± 28.36 optical density (OD) vs 95 ± 17.40 OD, P < 0.0001]. Quantitative morphometry carried out by using immuno-gold detection at transmission electron microscopy confirms an increased PrPc expression in PDAC ductal cells of all patients and allows to detect a specific compartmentalization of PrPc within tumor cells. In particular, the number of immuno-gold particles of PrPc was significantly higher in PDAC cells respect to controls, when considering the whole cell (19.8 ± 0.79 particles vs 9.44 ± 0.45, P < 0.0001). Remarkably, considering PDAC cells, the increase of PrPc was higher in the nucleus than cytosol of tumor cells, which indicates a shift in PrPc compartmentalization within tumor cells. In fact, the increase of immuno-gold within nuclear compartment exceeds at large the augment of PrPc which was detected in the cytosol (nucleus: 12.88 ± 0.59 particles vs 5.12 ± 0.32, P < 0.0001; cytosol: 7.74. ± 0.44 particles vs 4.3 ± 0.24, P < 0.0001). In order to analyze the prognostic impact of PrPc, we found a correlation between PrPc expression and cancer stage according to pathology results, with a significantly higher expression of PrPc for advanced stages. Moreover, 24 patients with a mean follow-up of 16.8 mo were considered. Immuno-blot analysis revealed a significantly higher expression of PrPc in patients with disease recurrence at 12 mo after radical surgery (360.71 ± 69.01 OD vs 170.23 ± 23.06 OD, P = 0.023), also in the subgroup of patients treated with adjuvant CT (368.36 ± 79.26 OD in the recurrence group vs 162.86 ± 24.16 OD, P = 0.028), which indicates a correlation with a higher chemo-resistance. CONCLUSION Expression of PrPc is significantly higher in PDAC cells compared with control, with the protein mainly placed in the nucleus. Preliminary clinical data confirm the correlation with a poorer prognosis.
Collapse
Affiliation(s)
- Matteo Bianchini
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Maria Anita Giambelluca
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Maria Concetta Scavuzzo
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Gregorio Di Franco
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Simone Guadagni
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Matteo Palmeri
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Niccolò Furbetta
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Desirée Gianardi
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Niccola Funel
- Division of Surgical Pathology, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa 56124, Italy
| | - Claudio Ricci
- Division of Surgical Pathology, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa 56124, Italy
| | - Raffaele Gaeta
- Division of Surgical Pathology, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa 56124, Italy
| | - Luca Emanuele Pollina
- Division of Surgical Pathology, Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa 56124, Italy
| | - Alfredo Falcone
- Division of Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Caterina Vivaldi
- Division of Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Giulio Di Candio
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
| | - Francesca Biagioni
- IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli 86077, Italy
| | | | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
- EndoCAS (Center for Computer Assisted Surgery), University of Pisa, Pisa 56124, Italy
| | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56124, Italy
- IRCCS Neuromed, Istituto Neurologico Mediterraneo, Pozzilli 86077, Italy
| |
Collapse
|
8
|
With or without You: Co-Chaperones Mediate Health and Disease by Modifying Chaperone Function and Protein Triage. Cells 2021; 10:cells10113121. [PMID: 34831344 PMCID: PMC8619055 DOI: 10.3390/cells10113121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Heat shock proteins (HSPs) are a family of molecular chaperones that regulate essential protein refolding and triage decisions to maintain protein homeostasis. Numerous co-chaperone proteins directly interact and modify the function of HSPs, and these interactions impact the outcome of protein triage, impacting everything from structural proteins to cell signaling mediators. The chaperone/co-chaperone machinery protects against various stressors to ensure cellular function in the face of stress. However, coding mutations, expression changes, and post-translational modifications of the chaperone/co-chaperone machinery can alter the cellular stress response. Importantly, these dysfunctions appear to contribute to numerous human diseases. Therapeutic targeting of chaperones is an attractive but challenging approach due to the vast functions of HSPs, likely contributing to the off-target effects of these therapies. Current efforts focus on targeting co-chaperones to develop precise treatments for numerous diseases caused by defects in protein quality control. This review focuses on the recent developments regarding selected HSP70/HSP90 co-chaperones, with a concentration on cardioprotection, neuroprotection, cancer, and autoimmune diseases. We also discuss therapeutic approaches that highlight both the utility and challenges of targeting co-chaperones.
Collapse
|
9
|
Marques CMS, Pedron T, Batista BL, Cerchiaro G. Cellular prion protein activates Caspase 3 for apoptotic defense mechanism in astrocytes. Mol Cell Biochem 2021; 476:2149-2158. [PMID: 33547547 DOI: 10.1007/s11010-021-04078-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/25/2021] [Indexed: 12/31/2022]
Abstract
The cellular prion protein (PrPC) is anchored in the plasma membrane of cells, and it is highly present in cells of brain tissue, exerting numerous cellular and cognitive functions. The present study proves the importance of PrPC in the cellular defense mechanism and metal homeostasis in astrocytes cells. Through experimental studies using cell lines of immortalized mice astrocytes (wild type and knockout for PrPC), we showed that PrPc is involved in the apoptosis cell death process by the activation of Caspase 3, downregulation of p53, and cell cycle maintenance. Metal homeostasis was determined by inductively coupled plasma mass spectrometry technique, indicating the crucial role of PrPC to lower intracellular calcium. The lowered calcium concentration and the Caspase 3 downregulation in the PrPC-null astrocytes resulted in a faster growth rate in cells, comparing with PrPC wild-type one. The presence of PrPC shows to be essential to cell death and healthy growth. In conclusion, our results show for the first time that astrocyte knockout cells for the cellular prion protein could modulate apoptosis-dependent cell death pathways.
Collapse
Affiliation(s)
- Caroline M S Marques
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André, SP, 09210-580, Brazil
| | - Tatiana Pedron
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André, SP, 09210-580, Brazil
| | - Bruno L Batista
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André, SP, 09210-580, Brazil
| | - Giselle Cerchiaro
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André, SP, 09210-580, Brazil.
| |
Collapse
|
10
|
The Role of Cellular Prion Protein in Promoting Stemness and Differentiation in Cancer. Cancers (Basel) 2021; 13:cancers13020170. [PMID: 33418999 PMCID: PMC7825291 DOI: 10.3390/cancers13020170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Aside from its well-established role in prion disorders, in the last decades the significance of cellular prion protein (PrPC) expression in human cancers has attracted great attention. An extensive body of work provided evidence that PrPC contributes to tumorigenesis by regulating tumor growth, differentiation, and resistance to conventional therapies. In particular, PrPC over-expression has been related to the acquisition of a malignant phenotype of cancer stem cells (CSCs) in a variety of solid tumors, encompassing pancreatic ductal adenocarcinoma, osteosarcoma, breast, gastric, and colorectal cancers, and primary brain tumors as well. According to consensus, increased levels of PrPC endow CSCs with self-renewal, proliferative, migratory, and invasive capacities, along with increased resistance to anti-cancer agents. In addition, increasing evidence demonstrates that PrPc also participates in multi-protein complexes to modulate the oncogenic properties of CSCs, thus sustaining tumorigenesis. Therefore, strategies aimed at targeting PrPC and/or PrPC-organized complexes could be a promising approach for anti-cancer therapy. Abstract Cellular prion protein (PrPC) is seminal to modulate a variety of baseline cell functions to grant homeostasis. The classic role of such a protein was defined as a chaperone-like molecule being able to rescue cell survival. Nonetheless, PrPC also represents the precursor of the deleterious misfolded variant known as scrapie prion protein (PrPSc). This variant is detrimental in a variety of prion disorders. This multi-faceted role of PrP is greatly increased by recent findings showing how PrPC in its folded conformation may foster tumor progression by acting at multiple levels. The present review focuses on such a cancer-promoting effect. The manuscript analyzes recent findings on the occurrence of PrPC in various cancers and discusses the multiple effects, which sustain cancer progression. Within this frame, the effects of PrPC on stemness and differentiation are discussed. A special emphasis is provided on the spreading of PrPC and the epigenetic effects, which are induced in neighboring cells to activate cancer-related genes. These detrimental effects are further discussed in relation to the aberrancy of its physiological and beneficial role on cell homeostasis. A specific paragraph is dedicated to the role of PrPC beyond its effects in the biology of cancer to represent a potential biomarker in the follow up of patients following surgical resection.
Collapse
|
11
|
Prado MB, Melo Escobar MI, Alves RN, Coelho BP, Fernandes CFDL, Boccacino JM, Iglesia RP, Lopes MH. Prion Protein at the Leading Edge: Its Role in Cell Motility. Int J Mol Sci 2020; 21:E6677. [PMID: 32932634 PMCID: PMC7555277 DOI: 10.3390/ijms21186677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Cell motility is a central process involved in fundamental biological phenomena during embryonic development, wound healing, immune surveillance, and cancer spreading. Cell movement is complex and dynamic and requires the coordinated activity of cytoskeletal, membrane, adhesion and extracellular proteins. Cellular prion protein (PrPC) has been implicated in distinct aspects of cell motility, including axonal growth, transendothelial migration, epithelial-mesenchymal transition, formation of lamellipodia, and tumor migration and invasion. The preferential location of PrPC on cell membrane favors its function as a pivotal molecule in cell motile phenotype, being able to serve as a scaffold protein for extracellular matrix proteins, cell surface receptors, and cytoskeletal multiprotein complexes to modulate their activities in cellular movement. Evidence points to PrPC mediating interactions of multiple key elements of cell motility at the intra- and extracellular levels, such as integrins and matrix proteins, also regulating cell adhesion molecule stability and cell adhesion cytoskeleton dynamics. Understanding the molecular mechanisms that govern cell motility is critical for tissue homeostasis, since uncontrolled cell movement results in pathological conditions such as developmental diseases and tumor dissemination. In this review, we discuss the relevant contribution of PrPC in several aspects of cell motility, unveiling new insights into both PrPC function and mechanism in a multifaceted manner either in physiological or pathological contexts.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marilene Hohmuth Lopes
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.B.P.); (M.I.M.E.); (R.N.A.); (B.P.C.); (C.F.d.L.F.); (J.M.B.); (R.P.I.)
| |
Collapse
|
12
|
Bianchini M, Giambelluca MA, Scavuzzo MC, Di Franco G, Guadagni S, Palmeri M, Furbetta N, Gianardi D, Funel N, Pollina LE, Di Candio G, Fornai F, Morelli L. The occurrence of prion protein in surgically resected pancreatic adenocarcinoma. Pancreatology 2020; 20:1218-1225. [PMID: 32828686 DOI: 10.1016/j.pan.2020.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/24/2020] [Accepted: 08/10/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Among the several new targets for the comprehension of the biology of pancreatic ductal adenocarcinoma (PDAC), Prion proteins (PrPc) deserve particular mention, since they share a marked neurotropism. Actually, PrPc could have also a role in tumorigenesis, as recently demonstrated. However, only few in vitro studies in cell cultures showed the occurrence of PrPc in PDAC cells. We aim to evaluate the presence of PrPc in vivo in PDAC tissues as a potential new biomarker. METHODS Samples from tumors of 23 patients undergone pancreatic resections from July 2018 to May 2020 at our institution were collected and analyzed. Immunohistochemistry and western blotting of PDAC tissues were compared with control tissues. Immunohistochemistry was used also to evaluate the localization of PrPc and of CD155, a tumoral stem-cell marker. RESULTS All cases were moderately differentiated PDAC, with perineural invasion (PNI) in 19/23 cases (83%). According to western-blot analysis, PrPc was markedly expressed in PDAC tissues (273.5 ± 44.63 OD) respect to controls (100 ± 28.35 OD, p = 0.0018). Immunohistochemistry confirmed these findings, with higher linear staining of PrPc in PDAC ducts (127.145 ± 7.56 μm vs 75.21 ± 5.01 μm, p < 0.0001). PrPc and CD155 exactly overlapped in ductal tumoral cells, highlighting the possible relationship of PrPc with cancer stemness. Finally, PrPc expression related with cancer stage and there was a potential correspondence with PNI. CONCLUSIONS Our work provides evidence for increased levels of PrPc in PDAC. This might contribute to cancer aggressiveness and provides a potentially new biomarker. Work is in progress to decipher clinical implications.
Collapse
Affiliation(s)
- Matteo Bianchini
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Maria Anita Giambelluca
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Maria Concetta Scavuzzo
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Gregorio Di Franco
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Simone Guadagni
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Matteo Palmeri
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Niccolò Furbetta
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Desirée Gianardi
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Niccola Funel
- Division of Surgical Pathology, Department of Surgical, Medical Molecular Pathology and Critical Area, University of Pisa, 56124, Pisa, Italy
| | - Luca Emanuele Pollina
- Division of Surgical Pathology, Department of Surgical, Medical Molecular Pathology and Critical Area, University of Pisa, 56124, Pisa, Italy
| | - Giulio Di Candio
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy
| | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy; IRCCS Neuromed - Istituto Neurologico Mediterraneo, 86077, Pozzilli, Italy
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124, Pisa, Italy; EndoCAS (Center for Computer Assisted Surgery), University of Pisa, 56124, Pisa, Italy.
| |
Collapse
|
13
|
Lackie RE, Marques-Lopes J, Ostapchenko VG, Good S, Choy WY, van Oosten-Hawle P, Pasternak SH, Prado VF, Prado MAM. Increased levels of Stress-inducible phosphoprotein-1 accelerates amyloid-β deposition in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2020; 8:143. [PMID: 32825842 PMCID: PMC7441634 DOI: 10.1186/s40478-020-01013-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 02/08/2023] Open
Abstract
Molecular chaperones and co-chaperones, which are part of the protein quality control machinery, have been shown to regulate distinct aspects of Alzheimer's Disease (AD) pathology in multiple ways. Notably, the co-chaperone STI1, which presents increased levels in AD, can protect mammalian neurons from amyloid-β toxicity in vitro and reduced STI1 levels worsen Aβ toxicity in C. elegans. However, whether increased STI1 levels can protect neurons in vivo remains unknown. We determined that overexpression of STI1 and/or Hsp90 protected C. elegans expressing Aβ(3-42) against Aβ-mediated paralysis. Mammalian neurons were also protected by elevated levels of endogenous STI1 in vitro, and this effect was mainly due to extracellular STI1. Surprisingly, in the 5xFAD mouse model of AD, by overexpressing STI1, we find increased amyloid burden, which amplifies neurotoxicity and worsens spatial memory deficits in these mutants. Increased levels of STI1 disturbed the expression of Aβ-regulating enzymes (BACE1 and MMP-2), suggesting potential mechanisms by which amyloid burden is increased in mice. Notably, we observed that STI1 accumulates in dense-core AD plaques in both 5xFAD mice and human brain tissue. Our findings suggest that elevated levels of STI1 contribute to Aβ accumulation, and that STI1 is deposited in AD plaques in mice and humans. We conclude that despite the protective effects of STI1 in C. elegans and in mammalian cultured neurons, in vivo, the predominant effect of elevated STI1 is deleterious in AD.
Collapse
Affiliation(s)
- Rachel E Lackie
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada
| | - Jose Marques-Lopes
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
| | - Valeriy G Ostapchenko
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
| | - Sarah Good
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Wing-Yiu Choy
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, Medical Sciences Building, 1151 Richmond St. N, London, N6A 5B7, Canada
| | - Patricija van Oosten-Hawle
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stephen H Pasternak
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
- St. Joseph's Health Care London-Parkwood Institute, St. Joseph's Hospital, 268 Grosvenor St Room A1-015, London, N6A 4V2, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, 1151 Richmond St, London, N6A 3K7, Canada
| | - Vania F Prado
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada.
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Anatomy & Cell Biology, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Ontario, Canada.
| | - Marco A M Prado
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada.
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Anatomy & Cell Biology, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Ontario, Canada.
| |
Collapse
|
14
|
Puig B, Yang D, Brenna S, Altmeppen HC, Magnus T. Show Me Your Friends and I Tell You Who You Are: The Many Facets of Prion Protein in Stroke. Cells 2020; 9:E1609. [PMID: 32630841 PMCID: PMC7407975 DOI: 10.3390/cells9071609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke belongs to the leading causes of mortality and disability worldwide. Although treatments for the acute phase of stroke are available, not all patients are eligible. There is a need to search for therapeutic options to promote neurological recovery after stroke. The cellular prion protein (PrPC) has been consistently linked to a neuroprotective role after ischemic damage: it is upregulated in the penumbra area following stroke in humans, and animal models of stroke have shown that lack of PrPC aggravates the ischemic damage and lessens the functional outcome. Mechanistically, these effects can be linked to numerous functions attributed to PrPC: (1) as a signaling partner of the PI3K/Akt and MAPK pathways, (2) as a regulator of glutamate receptors, and (3) promoting stem cell homing mechanisms, leading to angio- and neurogenesis. PrPC can be cleaved at different sites and the proteolytic fragments can account for the manifold functions. Moreover, PrPC is present on extracellular vesicles (EVs), released membrane particles originating from all types of cells that have drawn attention as potential therapeutic tools in stroke and many other diseases. Thus, identification of the many mechanisms underlying PrPC-induced neuroprotection will not only provide further understanding of the physiological functions of PrPC but also new ideas for possible treatment options after ischemic stroke.
Collapse
Affiliation(s)
- Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | - Denise Yang
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | | | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| |
Collapse
|
15
|
da Fonseca ACC, Matias D, Geraldo LHM, Leser FS, Pagnoncelli I, Garcia C, do Amaral RF, da Rosa BG, Grimaldi I, de Camargo Magalhães ES, Cóppola-Segovia V, de Azevedo EM, Zanata SM, Lima FRS. The multiple functions of the co-chaperone stress inducible protein 1. Cytokine Growth Factor Rev 2020; 57:73-84. [PMID: 32561134 DOI: 10.1016/j.cytogfr.2020.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/22/2020] [Accepted: 06/02/2020] [Indexed: 12/18/2022]
Abstract
Stress inducible protein 1 (STI1) is a co-chaperone acting with Hsp70 and Hsp90 for the correct client proteins' folding and therefore for the maintenance of cellular homeostasis. Besides being expressed in the cytosol, STI1 can also be found both in the cell membrane and the extracellular medium playing several relevant roles in the central nervous system (CNS) and tumor microenvironment. During CNS development, in association with cellular prion protein (PrPc), STI1 regulates crucial events such as neuroprotection, neuritogenesis, astrocyte differentiation and survival. In cancer, STI1 is involved with tumor growth and invasion, is undoubtedly a pro-tumor factor, being considered as a biomarker and possibly therapeutic target for several malignancies. In this review, we discuss current knowledge and new findings on STI1 function as well as its role in tissue homeostasis, CNS and tumor progression.
Collapse
Affiliation(s)
| | - Diana Matias
- Molecular Bionics Laboratory, Department of Chemistry, University College London, London, WC1H 0AJ, United Kingdom
| | - Luiz Henrique Medeiros Geraldo
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil; Université de Paris, PARCC, INSERM, Paris, 75015, France
| | - Felipe Saceanu Leser
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil
| | - Iohana Pagnoncelli
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil
| | - Celina Garcia
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil
| | - Rackele Ferreira do Amaral
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil
| | - Barbara Gomes da Rosa
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil
| | - Izabella Grimaldi
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil
| | - Eduardo Sabino de Camargo Magalhães
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil; European Research Institute for the Biology of Aging, University of Groningen, Groningen, 9713 AV, Netherlands
| | - Valentín Cóppola-Segovia
- Departments of Basic Pathology and Cell Biology, Federal University of Paraná, Paraná, RJ, 81531-970, Brazil
| | - Evellyn Mayla de Azevedo
- Departments of Basic Pathology and Cell Biology, Federal University of Paraná, Paraná, RJ, 81531-970, Brazil
| | - Silvio Marques Zanata
- Departments of Basic Pathology and Cell Biology, Federal University of Paraná, Paraná, RJ, 81531-970, Brazil
| | - Flavia Regina Souza Lima
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-590, Brazil.
| |
Collapse
|
16
|
Thellung S, Corsaro A, Bosio AG, Zambito M, Barbieri F, Mazzanti M, Florio T. Emerging Role of Cellular Prion Protein in the Maintenance and Expansion of Glioma Stem Cells. Cells 2019; 8:cells8111458. [PMID: 31752162 PMCID: PMC6912268 DOI: 10.3390/cells8111458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
Cellular prion protein (PrPC) is a membrane-anchored glycoprotein representing the physiological counterpart of PrP scrapie (PrPSc), which plays a pathogenetic role in prion diseases. Relatively little information is however available about physiological role of PrPC. Although PrPC ablation in mice does not induce lethal phenotypes, impairment of neuronal and bone marrow plasticity was reported in embryos and adult animals. In neurons, PrPC stimulates neurite growth, prevents oxidative stress-dependent cell death, and favors antiapoptotic signaling. However, PrPC activity is not restricted to post-mitotic neurons, but promotes cell proliferation and migration during embryogenesis and tissue regeneration in adult. PrPC acts as scaffold to stabilize the binding between different membrane receptors, growth factors, and basement proteins, contributing to tumorigenesis. Indeed, ablation of PrPC expression reduces cancer cell proliferation and migration and restores cell sensitivity to chemotherapy. Conversely, PrPC overexpression in cancer stem cells (CSCs) from different tumors, including gliomas—the most malignant brain tumors—is predictive for poor prognosis, and correlates with relapses. The mechanisms of the PrPC role in tumorigenesis and its molecular partners in this activity are the topic of the present review, with a particular focus on PrPC contribution to glioma CSCs multipotency, invasiveness, and tumorigenicity.
Collapse
Affiliation(s)
- Stefano Thellung
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Alessandro Corsaro
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Alessia G. Bosio
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Martina Zambito
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Federica Barbieri
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
| | - Michele Mazzanti
- Dipartimento di Bioscienze, Università di Milano, 20133 Milano, Italy
- Correspondence: (T.F.); (M.M.); Tel.: +39-01-0353-8806 (T.F.); +39-02-5031-4958 (M.M.)
| | - Tullio Florio
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica (CEBR), Università di Genova, 16132 Genova, Italy; (S.T.); (A.C.); (A.G.B.); (M.Z.); (F.B.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Correspondence: (T.F.); (M.M.); Tel.: +39-01-0353-8806 (T.F.); +39-02-5031-4958 (M.M.)
| |
Collapse
|
17
|
Lackie RE, Razzaq AR, Farhan SMK, Qiu LR, Moshitzky G, Beraldo FH, Lopes MH, Maciejewski A, Gros R, Fan J, Choy WY, Greenberg DS, Martins VR, Duennwald ML, Lerch JP, Soreq H, Prado VF, Prado MAM. Modulation of hippocampal neuronal resilience during aging by the Hsp70/Hsp90 co-chaperone STI1. J Neurochem 2019; 153:727-758. [PMID: 31562773 DOI: 10.1111/jnc.14882] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/22/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022]
Abstract
Chaperone networks are dysregulated with aging, but whether compromised Hsp70/Hsp90 chaperone function disturbs neuronal resilience is unknown. Stress-inducible phosphoprotein 1 (STI1; STIP1; HOP) is a co-chaperone that simultaneously interacts with Hsp70 and Hsp90, but whose function in vivo remains poorly understood. We combined in-depth analysis of chaperone genes in human datasets, analysis of a neuronal cell line lacking STI1 and of a mouse line with a hypomorphic Stip1 allele to investigate the requirement for STI1 in aging. Our experiments revealed that dysfunctional STI1 activity compromised Hsp70/Hsp90 chaperone network and neuronal resilience. The levels of a set of Hsp90 co-chaperones and client proteins were selectively affected by reduced levels of STI1, suggesting that their stability depends on functional Hsp70/Hsp90 machinery. Analysis of human databases revealed a subset of co-chaperones, including STI1, whose loss of function is incompatible with life in mammals, albeit they are not essential in yeast. Importantly, mice expressing a hypomorphic STI1 allele presented spontaneous age-dependent hippocampal neurodegeneration and reduced hippocampal volume, with consequent spatial memory deficit. We suggest that impaired STI1 function compromises Hsp70/Hsp90 chaperone activity in mammals and can by itself cause age-dependent hippocampal neurodegeneration in mice. Cover Image for this issue: doi: 10.1111/jnc.14749.
Collapse
Affiliation(s)
- Rachel E Lackie
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Program in Neuroscience, University of Western Ontario, London, Ontario, Canada
| | - Abdul R Razzaq
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Program in Neuroscience, University of Western Ontario, London, Ontario, Canada
| | - Sali M K Farhan
- Analytic and Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, and The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Lily R Qiu
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Gilli Moshitzky
- Department of Biological Chemistry, The Edmond and Lily Safra Center for Brain Sciences, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Flavio H Beraldo
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Marilene H Lopes
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Laboratory of Neurobiology and Stem cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Andrzej Maciejewski
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Robert Gros
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Jue Fan
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Wing-Yiu Choy
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - David S Greenberg
- Department of Biological Chemistry, The Edmond and Lily Safra Center for Brain Sciences, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vilma R Martins
- International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Martin L Duennwald
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Hermona Soreq
- Department of Biological Chemistry, The Edmond and Lily Safra Center for Brain Sciences, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vania F Prado
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Program in Neuroscience, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Marco A M Prado
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Program in Neuroscience, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
18
|
Ryskalin L, Busceti CL, Biagioni F, Limanaqi F, Familiari P, Frati A, Fornai F. Prion Protein in Glioblastoma Multiforme. Int J Mol Sci 2019; 20:ijms20205107. [PMID: 31618844 PMCID: PMC6834196 DOI: 10.3390/ijms20205107] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022] Open
Abstract
The cellular prion protein (PrPc) is an evolutionarily conserved cell surface protein encoded by the PRNP gene. PrPc is ubiquitously expressed within nearly all mammalian cells, though most abundantly within the CNS. Besides being implicated in the pathogenesis and transmission of prion diseases, recent studies have demonstrated that PrPc contributes to tumorigenesis by regulating tumor growth, differentiation, and resistance to conventional therapies. In particular, PrPc over-expression has been related to the acquisition of a malignant phenotype of cancer stem cells (CSCs) in a variety of solid tumors, encompassing pancreatic ductal adenocarcinoma (PDAC), osteosarcoma, breast cancer, gastric cancer, and primary brain tumors, mostly glioblastoma multiforme (GBM). Thus, PrPc is emerging as a key in maintaining glioblastoma cancer stem cells’ (GSCs) phenotype, thereby strongly affecting GBM infiltration and relapse. In fact, PrPc contributes to GSCs niche’s maintenance by modulating GSCs’ stem cell-like properties while restraining them from differentiation. This is the first review that discusses the role of PrPc in GBM. The manuscript focuses on how PrPc may act on GSCs to modify their expression and translational profile while making the micro-environment surrounding the GSCs niche more favorable to GBM growth and infiltration.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy.
| | - Carla L Busceti
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy.
| | | | - Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy.
| | - Pietro Familiari
- Department of Neuroscience, Mental Health and Sense Organs NESMOS, Sapienza University of Rome, 00185 Rome, Italy.
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126 Pisa, Italy.
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli, Italy.
| |
Collapse
|
19
|
Lee YH, Lee HT, Chen CL, Chang CH, Hsu CY, Shyu WC. Role of FOXC1 in regulating APSCs self-renewal via STI-1/PrP C signaling. Am J Cancer Res 2019; 9:6443-6465. [PMID: 31588228 PMCID: PMC6771253 DOI: 10.7150/thno.35619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/20/2019] [Indexed: 01/03/2023] Open
Abstract
Forkhead box protein C1 (FOXC1) is known to regulate developmental processes in the skull and brain. Methods: The unique multipotent arachnoid-pia stem cells (APSCs) isolated from human and mouse arachnoid-pia membranes of meninges were grown as 3D spheres and displayed a capacity for self-renewal. Additionally, APSCs also expressed the surface antigens as mesenchymal stem cells. By applying the FOXC1 knockout mice and mouse brain explants, signaling cascade of FOXC1-STI-1-PrPC was investigated to demonstrate the molecular regulatory pathway for APSCs self-renewal. Moreover, APSCs implantation in stroke model was also verified whether neurogenic property of APSCs could repair the ischemic insult of the stroke brain. Results: Activated FOXC1 regulated the proliferation of APSCs in a cell cycle-dependent manner, whereas FOXC1-mediated APSCs self-renewal was abolished in FOXC1 knockout mice (FOXC1-/- mice). Moreover, upregulation of STI-1 regulated by FOXC1 enhanced cell survival and self-renewal of APSCs through autocrine signaling of cellular prion protein (PrPC). Mouse brain explants STI-1 rescues the cortical phenotype in vitro and induces neurogenesis in the FOXC1 -/- mouse brain. Furthermore, administration of APSCs in ischemic brain restored the neuroglial microenvironment and improved neurological dysfunction. Conclusion: We identified a novel role for FOXC1 in the direct regulation of the STI-1-PrPC signaling pathway to promote cell proliferation and self-renewal of APSCs.
Collapse
|
20
|
Matamoros-Angles A, Gayosso LM, Richaud-Patin Y, di Domenico A, Vergara C, Hervera A, Sousa A, Fernández-Borges N, Consiglio A, Gavín R, López de Maturana R, Ferrer I, López de Munain A, Raya Á, Castilla J, Sánchez-Pernaute R, Del Río JA. iPS Cell Cultures from a Gerstmann-Sträussler-Scheinker Patient with the Y218N PRNP Mutation Recapitulate tau Pathology. Mol Neurobiol 2018; 55:3033-3048. [PMID: 28466265 PMCID: PMC5842509 DOI: 10.1007/s12035-017-0506-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/21/2017] [Indexed: 01/20/2023]
Abstract
Gerstmann-Sträussler-Scheinker (GSS) syndrome is a fatal autosomal dominant neurodegenerative prionopathy clinically characterized by ataxia, spastic paraparesis, extrapyramidal signs and dementia. In some GSS familiar cases carrying point mutations in the PRNP gene, patients also showed comorbid tauopathy leading to mixed pathologies. In this study we developed an induced pluripotent stem (iPS) cell model derived from fibroblasts of a GSS patient harboring the Y218N PRNP mutation, as well as an age-matched healthy control. This particular PRNP mutation is unique with very few described cases. One of the cases presented neurofibrillary degeneration with relevant Tau hyperphosphorylation. Y218N iPS-derived cultures showed relevant astrogliosis, increased phospho-Tau, altered microtubule-associated transport and cell death. However, they failed to generate proteinase K-resistant prion. In this study we set out to test, for the first time, whether iPS cell-derived neurons could be used to investigate the appearance of disease-related phenotypes (i.e, tauopathy) identified in the GSS patient.
Collapse
Affiliation(s)
- Andreu Matamoros-Angles
- Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, E-08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Lucía Mayela Gayosso
- Stem cells and neural repair laboratory, Fundación Inbiomed, San Sebastian, Gipuzkoa, Spain
- Proteomics unit (Prion lab), CIC bioGUNE, Parque tecnológico de Bizkaia, 48160, Derio, Bizkaia, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia, Spain
| | - Yvonne Richaud-Patin
- Centre de Medicina Regenerativa de Barcelona, c/ Dr. Aiguader 88, 08003, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), Madrid, Spain
| | - Angelique di Domenico
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
- Dept. Patologia i Terapèutica Experimental, Universitat de Barcelona, Barcelona, Spain
| | - Cristina Vergara
- Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, E-08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
- Laboratory of Histology, Neuroanatomy and Neuropathology (CP 620), ULB Neuroscience Institute. Université Libre de Bruxelles, Faculty of Medicine, Brussels, Belgium
| | - Arnau Hervera
- Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, E-08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Amaya Sousa
- Stem cells and neural repair laboratory, Fundación Inbiomed, San Sebastian, Gipuzkoa, Spain
| | - Natalia Fernández-Borges
- Proteomics unit (Prion lab), CIC bioGUNE, Parque tecnológico de Bizkaia, 48160, Derio, Bizkaia, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia, Spain
- CISA-INIA, Center for Animal Health Research, Madrid, Spain
| | - Antonella Consiglio
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
- Dept. Patologia i Terapèutica Experimental, Universitat de Barcelona, Barcelona, Spain
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Rosalina Gavín
- Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, E-08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | | | - Isidro Ferrer
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
- Dept. Patologia i Terapèutica Experimental, Universitat de Barcelona, Barcelona, Spain
| | - Adolfo López de Munain
- Instituto Biodonostia-Hospital Universitario Donostia, San Sebastian, Gipuzkoa, Spain
- Neurosciences Department, University of the Basque Country UPV-EHU, Bilbao, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), San Sebastian, Gipuzkoa, Spain
| | - Ángel Raya
- Centre de Medicina Regenerativa de Barcelona, c/ Dr. Aiguader 88, 08003, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), Madrid, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| | - Joaquín Castilla
- Proteomics unit (Prion lab), CIC bioGUNE, Parque tecnológico de Bizkaia, 48160, Derio, Bizkaia, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia, Spain.
| | - Rosario Sánchez-Pernaute
- Stem cells and neural repair laboratory, Fundación Inbiomed, San Sebastian, Gipuzkoa, Spain.
- Andalusian Initiative for Advanced Therapies, Junta de Andalusia, Seville, Spain.
| | - José Antonio Del Río
- Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri Reixac 15-21, E-08028, Barcelona, Spain.
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
21
|
Cellular prion protein controls stem cell-like properties of human glioblastoma tumor-initiating cells. Oncotarget 2018; 7:38638-38657. [PMID: 27229535 PMCID: PMC5122417 DOI: 10.18632/oncotarget.9575] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/04/2016] [Indexed: 12/17/2022] Open
Abstract
Prion protein (PrPC) is a cell surface glycoprotein whose misfolding is responsible for prion diseases. Although its physiological role is not completely defined, several lines of evidence propose that PrPC is involved in self-renewal, pluripotency gene expression, proliferation and differentiation of neural stem cells. Moreover, PrPC regulates different biological functions in human tumors, including glioblastoma (GBM). We analyzed the role of PrPC in GBM cell pathogenicity focusing on tumor-initiating cells (TICs, or cancer stem cells, CSCs), the subpopulation responsible for development, progression and recurrence of most malignancies. Analyzing four GBM CSC-enriched cultures, we show that PrPC expression is directly correlated with the proliferation rate of the cells. To better define its role in CSC biology, we knocked-down PrPC expression in two of these GBM-derived CSC cultures by specific lentiviral-delivered shRNAs. We provide evidence that CSC proliferation rate, spherogenesis and in vivo tumorigenicity are significantly inhibited in PrPC down-regulated cells. Moreover, PrPC down-regulation caused loss of expression of the stemness and self-renewal markers (NANOG, Sox2) and the activation of differentiation pathways (i.e. increased GFAP expression). Our results suggest that PrPC controls the stemness properties of human GBM CSCs and that its down-regulation induces the acquisition of a more differentiated and less oncogenic phenotype.
Collapse
|
22
|
Wang J, You H, Qi J, Yang C, Ren Y, Cheng H. Autocrine and paracrine STIP1 signaling promote osteolytic bone metastasis in renal cell carcinoma. Oncotarget 2017; 8:17012-17026. [PMID: 28199984 PMCID: PMC5370018 DOI: 10.18632/oncotarget.15222] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 01/04/2017] [Indexed: 12/30/2022] Open
Abstract
Bone metastases are responsible for some of the most devastating complications of renal cell carcinoma (RCC). However, pro-metastatic factors leading to the highly osteolytic characteristics of RCC bone metastasis have barely been explored. We previously developed novel bone-seeking RCC cell lines by the in vivo selection strategy and performed a comparative proteome analysis on their total cell lysate. Here, we focused on STIP1 (stress-induced phosphoprotein 1), the high up-regulated protein in the bone-seeking cells, and explored its clinical relevance and functions in RCC bone metastasis. We observed high levels of both intracellular and extracellular STIP1 protein in bone metastatic tissue samples. Elevated STIP1 mRNA in the primary RCC tumors remarkably correlated with worse clinical outcomes. Furthermore, both human recombinant STIP1 protein and anti-STIP1 neutralizing antibody were used in the functional studies. We found that 1) STIP1 protein on the extracellular surface of tumor cells promoted the proliferation and migration/invasion of RCC tumor cells through the autocrine STIP1-ALK2-SMAD1/5 pathway; and 2) STIP1 protein secreted into the extracellular tumor stromal area, promoted the differentiation of osteoclasts through the paracrine STIP1-PrPc-ERK1/2 pathway. Increased cathepsin K (CTSK), the key enzyme secreted by osteoclasts to degrade collagen and other matrix proteins during bone resorption was further detected in the differentiated osteoclasts. These results provide evidence of the great potential of STIP1 as a novel biomarker and therapeutic target in RCC bone metastasis.
Collapse
Affiliation(s)
- Jiang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R.China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R.China
| | - Jun Qi
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R.China
| | - Caihong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R.China
| | - Ye Ren
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R.China
| | - Hao Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R.China
| |
Collapse
|
23
|
Hirsch TZ, Martin-Lannerée S, Mouillet-Richard S. Functions of the Prion Protein. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 150:1-34. [PMID: 28838656 DOI: 10.1016/bs.pmbts.2017.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although initially disregarded compared to prion pathogenesis, the functions exerted by the cellular prion protein PrPC have gained much interest over the past two decades. Research aiming at unraveling PrPC functions started to intensify when it became appreciated that it would give clues as to how it is subverted in the context of prion infection and, more recently, in the context of Alzheimer's disease. It must now be admitted that PrPC is implicated in an incredible variety of biological processes, including neuronal homeostasis, stem cell fate, protection against stress, or cell adhesion. It appears that these diverse roles can all be fulfilled through the involvement of PrPC in cell signaling events. Our aim here is to provide an overview of our current understanding of PrPC functions from the animal to the molecular scale and to highlight some of the remaining gaps that should be addressed in future research.
Collapse
Affiliation(s)
- Théo Z Hirsch
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Séverine Martin-Lannerée
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Sophie Mouillet-Richard
- INSERM UMR 1124, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France.
| |
Collapse
|
24
|
Miyakoshi LM, Marques-Coelho D, De Souza LER, Lima FRS, Martins VR, Zanata SM, Hedin-Pereira C. Evidence of a Cell Surface Role for Hsp90 Complex Proteins Mediating Neuroblast Migration in the Subventricular Zone. Front Cell Neurosci 2017; 11:138. [PMID: 28567003 PMCID: PMC5434112 DOI: 10.3389/fncel.2017.00138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/26/2017] [Indexed: 11/30/2022] Open
Abstract
In most mammalian brains, the subventricular zone (SVZ) is a germinative layer that maintains neurogenic activity throughout adulthood. Neuronal precursors arising from this region migrate through the rostral migratory stream (RMS) and reach the olfactory bulbs where they differentiate and integrate into the local circuitry. Recently, studies have shown that heat shock proteins have an important role in cancer cell migration and blocking Hsp90 function was shown to hinder cell migration in the developing cerebellum. In this work, we hypothesize that chaperone complexes may have an important function regulating migration of neuronal precursors from the subventricular zone. Proteins from the Hsp90 complex are present in the postnatal SVZ as well as in the RMS. Using an in vitro SVZ explant model, we have demonstrated the expression of Hsp90 and Hop/STI1 by migrating neuroblasts. Treatment with antibodies against Hsp90 and co-chaperone Hop/STI1, as well as Hsp90 and Hsp70 inhibitors hinder neuroblast chain migration. Time-lapse videomicroscopy analysis revealed that cell motility and average migratory speed was decreased after exposure to both antibodies and inhibitors. Antibodies recognizing Hsp90, Hsp70, and Hop/STI1 were found bound to the membranes of cells from primary SVZ cultures and biotinylation assays demonstrated that Hsp70 and Hop/STI1 could be found on the external leaflet of neuroblast membranes. The latter could also be detected in conditioned medium samples obtained from cultivated SVZ cells. Our results suggest that chaperones Hsp90, Hsp70, and co-chaperone Hop/STI1, components of the Hsp90 complex, regulate SVZ neuroblast migration in a concerted manner through an extracellular mechanism.
Collapse
Affiliation(s)
- Leo M Miyakoshi
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,Laboratory of Cellular NeuroAnatomy, Institute for Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Diego Marques-Coelho
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,Laboratory of Cellular NeuroAnatomy, Institute for Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Luiz E R De Souza
- Department of Basic Pathology, Federal University of ParanáParaná, Brazil
| | - Flavia R S Lima
- Institute for Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Vilma R Martins
- International Research Center, A.C. Camargo Cancer CenterSão Paulo, Brazil
| | - Silvio M Zanata
- Department of Basic Pathology, Federal University of ParanáParaná, Brazil
| | - Cecilia Hedin-Pereira
- Biophysics Institute Carlos Chagas Filho, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,Laboratory of Cellular NeuroAnatomy, Institute for Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil.,VPPLR-Fundação Oswaldo Cruz (Fiocruz)Rio de Janeiro, Brazil
| |
Collapse
|
25
|
Lackie RE, Maciejewski A, Ostapchenko VG, Marques-Lopes J, Choy WY, Duennwald ML, Prado VF, Prado MAM. The Hsp70/Hsp90 Chaperone Machinery in Neurodegenerative Diseases. Front Neurosci 2017; 11:254. [PMID: 28559789 PMCID: PMC5433227 DOI: 10.3389/fnins.2017.00254] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/20/2017] [Indexed: 12/12/2022] Open
Abstract
The accumulation of misfolded proteins in the human brain is one of the critical features of many neurodegenerative diseases, including Alzheimer's disease (AD). Assembles of beta-amyloid (Aβ) peptide—either soluble (oligomers) or insoluble (plaques) and of tau protein, which form neurofibrillary tangles, are the major hallmarks of AD. Chaperones and co-chaperones regulate protein folding and client maturation, but they also target misfolded or aggregated proteins for refolding or for degradation, mostly by the proteasome. They form an important line of defense against misfolded proteins and are part of the cellular quality control system. The heat shock protein (Hsp) family, particularly Hsp70 and Hsp90, plays a major part in this process and it is well-known to regulate protein misfolding in a variety of diseases, including tau levels and toxicity in AD. However, the role of Hsp90 in regulating protein misfolding is not yet fully understood. For example, knockdown of Hsp90 and its co-chaperones in a Caenorhabditis elegans model of Aβ misfolding leads to increased toxicity. On the other hand, the use of Hsp90 inhibitors in AD mouse models reduces Aβ toxicity, and normalizes synaptic function. Stress-inducible phosphoprotein 1 (STI1), an intracellular co-chaperone, mediates the transfer of clients from Hsp70 to Hsp90. Importantly, STI1 has been shown to regulate aggregation of amyloid-like proteins in yeast. In addition to its intracellular function, STI1 can be secreted by diverse cell types, including astrocytes and microglia and function as a neurotrophic ligand by triggering signaling via the cellular prion protein (PrPC). Extracellular STI1 can prevent Aβ toxic signaling by (i) interfering with Aβ binding to PrPC and (ii) triggering pro-survival signaling cascades. Interestingly, decreased levels of STI1 in C. elegans can also increase toxicity in an amyloid model. In this review, we will discuss the role of intracellular and extracellular STI1 and the Hsp70/Hsp90 chaperone network in mechanisms underlying protein misfolding in neurodegenerative diseases, with particular focus on AD.
Collapse
Affiliation(s)
- Rachel E Lackie
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada.,Program in Neuroscience, University of Western OntarioLondon, ON, Canada
| | - Andrzej Maciejewski
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada.,Department of Biochemistry, University of Western OntarioLondon, ON, Canada
| | - Valeriy G Ostapchenko
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada
| | - Jose Marques-Lopes
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada
| | - Wing-Yiu Choy
- Department of Biochemistry, University of Western OntarioLondon, ON, Canada
| | - Martin L Duennwald
- Department of Pathology and Laboratory Medicine, University of Western OntarioLondon, ON, Canada
| | - Vania F Prado
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada.,Program in Neuroscience, University of Western OntarioLondon, ON, Canada.,Department of Physiology and Pharmacology, University of Western OntarioLondon, ON, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, ON, Canada
| | - Marco A M Prado
- Molecular Medicine, Robarts Research Institute, University of Western OntarioLondon, ON, Canada.,Program in Neuroscience, University of Western OntarioLondon, ON, Canada.,Department of Physiology and Pharmacology, University of Western OntarioLondon, ON, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western OntarioLondon, ON, Canada
| |
Collapse
|
26
|
Macedo JA, Schrama D, Duarte I, Tavares E, Renaut J, Futschik ME, Rodrigues PM, Melo EP. Membrane-enriched proteome changes and prion protein expression during neural differentiation and in neuroblastoma cells. BMC Genomics 2017; 18:319. [PMID: 28431525 PMCID: PMC5401558 DOI: 10.1186/s12864-017-3694-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/08/2017] [Indexed: 01/12/2023] Open
Abstract
Background The function of the prion protein, involved in the so-called prion diseases, remains a subject of intense debate and the possibility that it works as a pleiotropic protein through the interaction with multiple membrane proteins is somehow supported by recent reports. Therefore, the use of proteomic and bioinformatics combined to uncover cellular processes occurring together with changes in the expression of the prion protein may provide further insight into the putative pleiotropic role of the prion protein. Results This study assessed the membrane-enriched proteome changes accompanying alterations in the expression of the prion protein. A 2D-DIGE approach was applied to two cell lines after prefractionation towards the membrane protein subset: an embryonic stem cell line and the PK1 subline of neuroblastoma cells which efficiently propagates prion infection. Several proteins were differentially abundant with the increased expression of the prion protein during neural differentiation of embryonic stem cells and with the knockdown of the prion protein in PK1 cells. The identity of around 20% of the differentially abundant proteins was obtained by tandem MS. The catalytic subunit A of succinate dehydrogenase, a key enzyme for the aerobic energy metabolism and redox homeostasis, showed a similar abundance trend as the prion protein in both proteomic experiments. A gene ontology analysis revealed “myelin sheath”, “organelle membrane” and “focal adhesion” associated proteins as the main cellular components, and “protein folding” and “ATPase activity” as the biological processes enriched in the first set of differentially abundant proteins. The known interactome of these differentially abundant proteins was customized to reveal four interactors with the prion protein, including two heat shock proteins and a protein disulfide isomerase. Conclusions Overall, our study shows that expression of the prion protein occurs concomitantly with changes in chaperone activity and cell-redox homeostasis, emphasizing the functional link between these cellular processes and the prion protein. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3694-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J A Macedo
- CBMR, Center for Biomedical Research, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - D Schrama
- CCMAR, Centre of Marine Sciences of Algarve, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - I Duarte
- CBMR, Center for Biomedical Research, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - E Tavares
- CBMR, Center for Biomedical Research, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - J Renaut
- LIST, Luxembourg Institute of Science and Technology, Belvaux, Luxembourg
| | - M E Futschik
- CCMAR, Centre of Marine Sciences of Algarve, University of Algarve, Campus de Gambelas, Faro, Portugal.,School of Biomedical & Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, UK
| | - P M Rodrigues
- CCMAR, Centre of Marine Sciences of Algarve, University of Algarve, Campus de Gambelas, Faro, Portugal
| | - E P Melo
- CBMR, Center for Biomedical Research, University of Algarve, Campus de Gambelas, Faro, Portugal.
| |
Collapse
|
27
|
Iglesia RP, Prado MB, Cruz L, Martins VR, Santos TG, Lopes MH. Engagement of cellular prion protein with the co-chaperone Hsp70/90 organizing protein regulates the proliferation of glioblastoma stem-like cells. Stem Cell Res Ther 2017; 8:76. [PMID: 28412969 PMCID: PMC5392955 DOI: 10.1186/s13287-017-0518-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 02/10/2017] [Accepted: 02/21/2017] [Indexed: 12/26/2022] Open
Abstract
Background Glioblastoma (GBM), a highly aggressive brain tumor, contains a subpopulation of glioblastoma stem-like cells (GSCs) that play roles in tumor maintenance, invasion, and therapeutic resistance. GSCs are therefore a promising target for GBM treatment. Our group identified the cellular prion protein (PrPC) and its partner, the co-chaperone Hsp70/90 organizing protein (HOP), as potential target candidates due to their role in GBM tumorigenesis and in neural stem cell maintenance. Methods GSCs expressing different levels of PrPC were cultured as neurospheres with growth factors, and characterized with stem cells markers and adhesion molecules markers through immunofluorescence and flow cytometry. We than evaluated GSC self-renewal and proliferation by clonal density assays and BrdU incorporation, respectively, in front of recombinant HOP treatment, combined or not with a HOP peptide which mimics the PrPC binding site. Stable silencing of HOP was also performed in parental and/or PrPC-depleted cell populations, and proliferation in vitro and tumor growth in vivo were evaluated. Migration assays were performed on laminin-1 pre-coated glass. Results We observed that, when GBM cells are cultured as neurospheres, they express specific stemness markers such as CD133, CD15, Oct4, and SOX2; PrPC is upregulated compared to monolayer culture and co-localizes with CD133. PrPC silencing downregulates the expression of molecules associated with cancer stem cells, upregulates markers of cell differentiation and affects GSC self-renewal, pointing to a pivotal role for PrPC in the maintenance of GSCs. Exogenous HOP treatment increases proliferation and self-renewal of GSCs in a PrPC-dependent manner while HOP knockdown disturbs the proliferation process. In vivo, PrPC and/or HOP knockdown potently inhibits the growth of subcutaneously implanted glioblastoma cells. In addition, disruption of the PrPC-HOP complex by a HOP peptide, which mimics the PrPC binding site, affects GSC self-renewal and proliferation indicating that the HOP-PrPC complex is required for GSC stemness. Furthermore, PrPC-depleted GSCs downregulate cell adhesion-related proteins and impair cell migration indicating a putative role for PrPC in the cell surface stability of cell adhesion molecules and GBM cell invasiveness, respectively. Conclusions In conclusion, our results show that the modulation of HOP-PrPC engagement or the decrease of PrPC and HOP expression may represent a potential therapeutic intervention in GBM, regulating glioblastoma stem-like cell self-renewal, proliferation, and migration. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0518-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rebeca Piatniczka Iglesia
- Laboratory of Neurobiology and Stem cells, Department of Cell and Developmental Biology; Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524 - Cidade Universitária "Armando Salles Oliveira", Butanta - Sao Paulo, SP, 05508-000, Brazil
| | - Mariana Brandão Prado
- Laboratory of Neurobiology and Stem cells, Department of Cell and Developmental Biology; Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524 - Cidade Universitária "Armando Salles Oliveira", Butanta - Sao Paulo, SP, 05508-000, Brazil
| | - Lilian Cruz
- Laboratory of Neurobiology and Stem cells, Department of Cell and Developmental Biology; Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524 - Cidade Universitária "Armando Salles Oliveira", Butanta - Sao Paulo, SP, 05508-000, Brazil
| | - Vilma Regina Martins
- Laboratory of Cell and Molecular Biology, International Research Center, A.C. Camargo Cancer Center, Sao Paulo, SP, 02056-070, Brazil
| | - Tiago Góss Santos
- Laboratory of Cell and Molecular Biology, International Research Center, A.C. Camargo Cancer Center, Sao Paulo, SP, 02056-070, Brazil
| | - Marilene Hohmuth Lopes
- Laboratory of Neurobiology and Stem cells, Department of Cell and Developmental Biology; Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 1524 - Cidade Universitária "Armando Salles Oliveira", Butanta - Sao Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
28
|
Linden R. The Biological Function of the Prion Protein: A Cell Surface Scaffold of Signaling Modules. Front Mol Neurosci 2017; 10:77. [PMID: 28373833 PMCID: PMC5357658 DOI: 10.3389/fnmol.2017.00077] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 03/06/2017] [Indexed: 12/18/2022] Open
Abstract
The prion glycoprotein (PrPC) is mostly located at the cell surface, tethered to the plasma membrane through a glycosyl-phosphatydil inositol (GPI) anchor. Misfolding of PrPC is associated with the transmissible spongiform encephalopathies (TSEs), whereas its normal conformer serves as a receptor for oligomers of the β-amyloid peptide, which play a major role in the pathogenesis of Alzheimer’s Disease (AD). PrPC is highly expressed in both the nervous and immune systems, as well as in other organs, but its functions are controversial. Extensive experimental work disclosed multiple physiological roles of PrPC at the molecular, cellular and systemic levels, affecting the homeostasis of copper, neuroprotection, stem cell renewal and memory mechanisms, among others. Often each such process has been heralded as the bona fide function of PrPC, despite restricted attention paid to a selected phenotypic trait, associated with either modulation of gene expression or to the engagement of PrPC with a single ligand. In contrast, the GPI-anchored prion protein was shown to bind several extracellular and transmembrane ligands, which are required to endow that protein with the ability to play various roles in transmembrane signal transduction. In addition, differing sets of those ligands are available in cell type- and context-dependent scenarios. To account for such properties, we proposed that PrPC serves as a dynamic platform for the assembly of signaling modules at the cell surface, with widespread consequences for both physiology and behavior. The current review advances the hypothesis that the biological function of the prion protein is that of a cell surface scaffold protein, based on the striking similarities of its functional properties with those of scaffold proteins involved in the organization of intracellular signal transduction pathways. Those properties are: the ability to recruit spatially restricted sets of binding molecules involved in specific signaling; mediation of the crosstalk of signaling pathways; reciprocal allosteric regulation with binding partners; compartmentalized responses; dependence of signaling properties upon posttranslational modification; and stoichiometric requirements and/or oligomerization-dependent impact on signaling. The scaffold concept may contribute to novel approaches to the development of effective treatments to hitherto incurable neurodegenerative diseases, through informed modulation of prion protein-ligand interactions.
Collapse
Affiliation(s)
- Rafael Linden
- Laboratory of Neurogenesis, Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
29
|
Wu ZQ, Li D, Huang Y, Chen XP, Huang W, Liu CF, Zhao HQ, Xu RX, Cheng M, Schachner M, Ma QH. Caspr Controls the Temporal Specification of Neural Progenitor Cells through Notch Signaling in the Developing Mouse Cerebral Cortex. Cereb Cortex 2017; 27:1369-1385. [PMID: 26740489 DOI: 10.1093/cercor/bhv318] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The generation of layer-specific neurons and astrocytes by radial glial cells during development of the cerebral cortex follows a precise temporal sequence, which is regulated by intrinsic and extrinsic factors. The molecular mechanisms controlling the timely generation of layer-specific neurons and astrocytes remain not fully understood. In this study, we show that the adhesion molecule contactin-associated protein (Caspr), which is involved in the maintenance of the polarized domains of myelinated axons, is essential for the timing of generation of neurons and astrocytes in the developing mouse cerebral cortex. Caspr is expressed by radial glial cells, which are neural progenitor cells that generate both neurons and astrocytes. Absence of Caspr in neural progenitor cells delays the production cortical neurons and induces precocious formation of cortical astrocytes, without affecting the numbers of progenitor cells. At the molecular level, Caspr cooperates with the intracellular domain of Notch to repress transcription of the Notch effector Hes1. Suppression of Notch signaling via a Hes1 shRNA rescues the abnormal neurogenesis and astrogenesis in Caspr-deficient mice. These findings establish Caspr as a novel key regulator that controls the temporal specification of cell fate in radial glial cells of the developing cerebral cortex through Notch signaling.
Collapse
Affiliation(s)
- Zhi-Qiang Wu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Di Li
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ya Huang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Xi-Ping Chen
- Department of Forensic Medicine, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Wenhui Huang
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg D-66421, Germany
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - He-Qing Zhao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ru-Xiang Xu
- Affiliated Bayi Brain Hospital, Beijing Military Hospital, Southern Medical University, Beijing 100070, China
| | - Mei Cheng
- Binzhou Medical University, Yantai, Shandong Province 264000, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| |
Collapse
|
30
|
Abstract
Traditional primary and secondary cell cultures have been used for the investigation of prion biology and disease for many years. While both types of cultures produce highly valid and immensely valuable results, they also have their limitations; traditional cell lines are often derived from cancers, therefore subject to numerous DNA changes, and primary cultures are labor-intensive and expensive to produce requiring sacrifice of many animals. Neural stem cell (NSC) cultures are a relatively new technology to be used for the study of prion biology and disease. While NSCs are subject to their own limitations-they are generally cultured ex vivo in environments that artificially force their growth-they also have their own unique advantages. NSCs retain the ability for self-renewal and can therefore be propagated in culture similarly to secondary cultures without genetic manipulation. In addition, NSCs are multipotent; they can be induced to differentiate into mature cells of central nervous system (CNS) linage. The combination of self-renewal and multipotency allows NSCs to be used as a primary cell line over multiple generations saving time, costs, and animal harvests, thus providing a valuable addition to the existing cell culture repertoire used for investigation of prion biology and disease. Furthermore, NSC cultures can be generated from mice of any genotype, either by embryonic harvest or harvest from adult brain, allowing gene expression to be studied without further genetic manipulation. This chapter describes a standard method of culturing adult NSCs and assays for monitoring NSC growth, migration, and differentiation and revisits basic reactive oxygen species detection in the context of NSC cultures.
Collapse
Affiliation(s)
- Cathryn L Haigh
- Department of Medicine, Melbourne Brain Centre, Royal Melbourne Hospital, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3010, Australia. .,Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|
31
|
Martin-Lannerée S, Halliez S, Hirsch TZ, Hernandez-Rapp J, Passet B, Tomkiewicz C, Villa-Diaz A, Torres JM, Launay JM, Béringue V, Vilotte JL, Mouillet-Richard S. The Cellular Prion Protein Controls Notch Signaling in Neural Stem/Progenitor Cells. Stem Cells 2016; 35:754-765. [PMID: 27641601 DOI: 10.1002/stem.2501] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 12/26/2022]
Abstract
The prion protein is infamous for its involvement in a group of neurodegenerative diseases known as Transmissible Spongiform Encephalopathies. In the longstanding quest to decipher the physiological function of its cellular isoform, PrPC , the discovery of its participation to the self-renewal of hematopoietic and neural stem cells has cast a new spotlight on its potential role in stem cell biology. However, still little is known on the cellular and molecular mechanisms at play. Here, by combining in vitro and in vivo murine models of PrPC depletion, we establish that PrPC deficiency severely affects the Notch pathway, which plays a major role in neural stem cell maintenance. We document that the absence of PrPC in a neuroepithelial cell line or in primary neurospheres is associated with drastically reduced expression of Notch ligands and receptors, resulting in decreased levels of Notch target genes. Similar alterations of the Notch pathway are recovered in the neuroepithelium of Prnp-/- embryos during a developmental window encompassing neural tube closure. In addition, in line with Notch defects, our data show that the absence of PrPC results in altered expression of Nestin and Olig2 as well as N-cadherin distribution. We further provide evidence that PrPC controls the expression of the epidermal growth factor receptor (EGFR) downstream from Notch. Finally, we unveil a negative feedback action of EGFR on both Notch and PrPC . As a whole, our study delineates a molecular scenario through which PrPC takes part to the self-renewal of neural stem and progenitor cells. Stem Cells 2017;35:754-765.
Collapse
Affiliation(s)
- Séverine Martin-Lannerée
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Sophie Halliez
- VIM, UR 892, INRA, Université Paris-Saclay, Jouy-en-Josas, France
| | - Théo Z Hirsch
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Julia Hernandez-Rapp
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Bruno Passet
- Department of Pharma Research, INRA UMR 1313, Génétique animale et biologie intégrative, Jouy-en-Josas, France
| | - Céline Tomkiewicz
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Ana Villa-Diaz
- Centro de Investigación en Sanidad Animal-INIA, U 942 Madrid, Spain
| | | | - Jean-Marie Launay
- AP-HP Service de Biochimie, Fondation FondaMental, INSERM U942 Hôpital Lariboisière, Paris, France.,Pharma Research Department, F. Hoffmann-La-Roche Ltd, Basel, Switzerland
| | - Vincent Béringue
- VIM, UR 892, INRA, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- Department of Pharma Research, INRA UMR 1313, Génétique animale et biologie intégrative, Jouy-en-Josas, France
| | - Sophie Mouillet-Richard
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| |
Collapse
|
32
|
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 2016; 483:1148-1155. [PMID: 27581199 DOI: 10.1016/j.bbrc.2016.07.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022]
Abstract
Prions are one of the few pathogens whose name is renowned at all population levels, after the dramatic years pervaded by the fear of eating prion-infected food. If now this, somehow irrational, scare of bovine meat inexorably transmitting devastating brain disorders is largely subdued, several prion-related issues are still unsolved, precluding the design of therapeutic approaches that could slow, if not halt, prion diseases. One unsolved issue is, for example, the role of the prion protein (PrPC), whole conformational misfolding originates the prion but whose physiologic reason d'etre in neurons, and in cells at large, remains enigmatic. Preceded by a historical outline, the present review will discuss the functional pleiotropicity ascribed to PrPC, and whether this aspect could fall, at least in part, into a more concise framework. It will also be devoted to radically different perspectives for PrPC, which have been recently brought to the attention of the scientific world with unexpected force. Finally, it will discuss the possible reasons allowing an evolutionary conserved and benign protein, as PrPC is, to turn into a high affinity receptor for pathologic misfolded oligomers, and to transmit their toxic message into neurons.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy.
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - M Catia Sorgato
- Department of Biomedical Sciences, University of Padova, Via Bassi 58/B, 35131 Padova, Italy; C.N.R. Institute of Neuroscience, University of Padova, Via Bassi 58/B, 35131 Padova, Italy.
| |
Collapse
|
33
|
Slapšak U, Salzano G, Amin L, Abskharon RNN, Ilc G, Zupančič B, Biljan I, Plavec J, Giachin G, Legname G. The N Terminus of the Prion Protein Mediates Functional Interactions with the Neuronal Cell Adhesion Molecule (NCAM) Fibronectin Domain. J Biol Chem 2016; 291:21857-21868. [PMID: 27535221 DOI: 10.1074/jbc.m116.743435] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Indexed: 12/22/2022] Open
Abstract
The cellular form of the prion protein (PrPC) is a highly conserved glycoprotein mostly expressed in the central and peripheral nervous systems by different cell types in mammals. A misfolded, pathogenic isoform, denoted as prion, is related to a class of neurodegenerative diseases known as transmissible spongiform encephalopathy. PrPC function has not been unequivocally clarified, and it is rather defined as a pleiotropic protein likely acting as a dynamic cell surface scaffolding protein for the assembly of different signaling modules. Among the variety of PrPC protein interactors, the neuronal cell adhesion molecule (NCAM) has been studied in vivo, but the structural basis of this functional interaction is still a matter of debate. Here we focused on the structural determinants responsible for human PrPC (HuPrP) and NCAM interaction using stimulated emission depletion (STED) nanoscopy, SPR, and NMR spectroscopy approaches. PrPC co-localizes with NCAM in mouse hippocampal neurons, and this interaction is mainly mediated by the intrinsically disordered PrPC N-terminal tail, which binds with high affinity to the NCAM fibronectin type-3 domain. NMR structural investigations revealed surface-interacting epitopes governing the interaction between HuPrP N terminus and the second module of the NCAM fibronectin type-3 domain. Our data provided molecular details about the interaction between HuPrP and the NCAM fibronectin domain, and revealed a new role of PrPC N terminus as a dynamic and functional element responsible for protein-protein interaction.
Collapse
Affiliation(s)
- Urška Slapšak
- From the Slovenian NMR Centre, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Giulia Salzano
- the Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, Trieste I-34136, Italy
| | - Ladan Amin
- the Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, Trieste I-34136, Italy
| | - Romany N N Abskharon
- the Structural Biology Research Center, Vrije Universiteit Brussel, VIB, Pleinlaan 2, 1050, Brussels, Belgium, the National Institute of Oceanography and Fisheries (NIOF), 11516 Cairo, Egypt, and the Van Andel Research Institute, Grand Rapids, Michigan 49503
| | - Gregor Ilc
- From the Slovenian NMR Centre, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia, the EN-FIST Centre of Excellence, Dunajska 156, SI-1000 Ljubljana, Slovenia
| | - Blaž Zupančič
- From the Slovenian NMR Centre, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
| | - Ivana Biljan
- the Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102A, Zagreb HR-10000, Croatia
| | - Janez Plavec
- From the Slovenian NMR Centre, National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia, the EN-FIST Centre of Excellence, Dunajska 156, SI-1000 Ljubljana, Slovenia, the Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia,
| | - Gabriele Giachin
- the Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, Trieste I-34136, Italy, the Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 Avenue des Martyrs, 38000-Grenoble, France
| | - Giuseppe Legname
- the Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, Trieste I-34136, Italy,
| |
Collapse
|
34
|
Baskakov IV, Katorcha E. Multifaceted Role of Sialylation in Prion Diseases. Front Neurosci 2016; 10:358. [PMID: 27551257 PMCID: PMC4976111 DOI: 10.3389/fnins.2016.00358] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/18/2016] [Indexed: 11/13/2022] Open
Abstract
Mammalian prion or PrP(Sc) is a proteinaceous infectious agent that consists of a misfolded, self-replicating state of a sialoglycoprotein called the prion protein, or PrP(C). Sialylation of the prion protein N-linked glycans was discovered more than 30 years ago, yet the role of sialylation in prion pathogenesis remains poorly understood. Recent years have witnessed extraordinary growth in interest in sialylation and established a critical role for sialic acids in host invasion and host-pathogen interactions. This review article summarizes current knowledge on the role of sialylation of the prion protein in prion diseases. First, we discuss the correlation between sialylation of PrP(Sc) glycans and prion infectivity and describe the factors that control sialylation of PrP(Sc). Second, we explain how glycan sialylation contributes to the prion replication barrier, defines strain-specific glycoform ratios, and imposes constraints for PrP(Sc) structure. Third, several topics, including a possible role for sialylation in animal-to-human prion transmission, prion lymphotropism, toxicity, strain interference, and normal function of PrP(C), are critically reviewed. Finally, a metabolic hypothesis on the role of sialylation in the etiology of sporadic prion diseases is proposed.
Collapse
Affiliation(s)
- Ilia V. Baskakov
- Department of Anatomy and Neurobiology, Center for Biomedical Engineering and Technology, University of Maryland School of MedicineBaltimore, MD, USA
| | | |
Collapse
|
35
|
de Lacerda TCS, Costa-Silva B, Giudice FS, Dias MVS, de Oliveira GP, Teixeira BL, Dos Santos TG, Martins VR. Prion protein binding to HOP modulates the migration and invasion of colorectal cancer cells. Clin Exp Metastasis 2016; 33:441-51. [PMID: 27112151 DOI: 10.1007/s10585-016-9788-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/29/2016] [Indexed: 12/19/2022]
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed malignancies. The generation of conventional treatments has improved, but approximately 50 % of patients with CRC who undergo potentially curative surgery ultimately relapse and die, usually as a consequence of metastatic disease. Our previous findings showed that engagement of the cellular prion protein (PrP(C)) to its ligand HSP70/90 heat shock organizing protein (HOP) induces proliferation of glioblastomas. In addition, PrP(C) has been described as an important modulator of colorectal tumor growth. Here, we investigated the biological relevance of the PrP(C)-HOP interaction in CRC cells. We demonstrate that HOP induced the migration and invasion of CRC cell lines in a PrP(C)-dependent manner and that phosphorylation of the ERK1/2 pathway is a downstream mediator of these effects. Additionally, we show that a HOP peptide with the ability to bind PrP(C) and abolish the PrP(C)-HOP interaction inhibited the migration and invasion of CRC cells. Together, these data indicate that the disruption of the PrP(C)-HOP complex could be a potential therapeutic target for modulating the migratory and invasive cellular properties that lead to metastatic CRC.
Collapse
Affiliation(s)
- Tonielli Cristina Sousa de Lacerda
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil.,Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), Av. Prof. Lineu Prestes, 748 - Cidade Universitária, São Paulo, SP, 05508-000, Brazil
| | - Bruno Costa-Silva
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil.,Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, 10021, USA.,Champalimaud Research, Champalimaud Centre for the Unknown, Av. Brasilia, Doca de Pedrouços, 1400-038, Lisbon, Portugal
| | - Fernanda Salgueiredo Giudice
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Marcos Vinicios Salles Dias
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Gabriela Pintar de Oliveira
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Bianca Luise Teixeira
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Tiago Goss Dos Santos
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil
| | - Vilma Regina Martins
- International Research Center, A.C.Camargo Cancer Center, Rua Taguá, 440 - Liberdade, São Paulo, SP, 01508-010, Brazil.
| |
Collapse
|
36
|
Abstract
In recent years, prion protein (PrP(C)) has been considered as a promising target molecule for cancer therapies, due its direct or indirect participation in tumor growth, metastasis, and resistance to cell death induced by chemotherapy. PrP(C) functions as a scaffold protein, forming multiprotein complexes on the plasma membrane, which elicits distinct signaling pathways involved in diverse biological phenomena and could be modulated depending on the cell type, complex composition, and organization. In addition, PrP(C) and its partners participate in self-renewal of embryonic, tissue-specific stem cells and cancer stem cells, which are suggested to be responsible for the origin, maintenance, relapse, and dissemination of tumors. Interference with protein-protein interaction has been recognized as an important therapeutic strategy in cancer; indeed, the possible interference in PrP(C) engagement with specific partners is a novel strategy. Recently, our group successfully used that approach to interfere with the interaction between PrP(C) and HSP-90/70 organizing protein (HOP, also known as stress-inducible protein 1 - STI1) to control the growth of human glioblastoma in animal models. Thus, PrP(C)-organized multicomplexes have emerged as feasible candidates for anti-tumor therapy, warranting further exploration.
Collapse
Affiliation(s)
- Tiago G Santos
- a International Research Center; AC Camargo Cancer Center ; São Paulo , Brazil
| | | | | |
Collapse
|
37
|
Bakkebø 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. [PMID: 26388873 PMCID: PMC4557099 DOI: 10.3389/fimmu.2015.00450] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/19/2015] [Indexed: 01/09/2023] Open
Abstract
Despite intensive studies since the 1990s, the physiological role of the cellular prion protein (PrP(C)) remains elusive. Here, we present a novel concept suggesting that PrP(C) contributes to immunological quiescence in addition to cell protection. PrP(C) is highly expressed in diverse organs that by multiple means are particularly protected from inflammation, such as the brain, eye, placenta, pregnant uterus, and testes, while at the same time it is expressed in most cells of the lymphoreticular system. In this paradigm, PrP(C) serves two principal roles: to modulate the inflammatory potential of immune cells and to protect vulnerable parenchymal cells against noxious insults generated through inflammation. Here, we review studies of PrP(C) physiology in view of this concept.
Collapse
Affiliation(s)
- Maren K. Bakkebø
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | | | - Arild Espenes
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Wilfred Goldmann
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Jörg Tatzelt
- Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Michael A. Tranulis
- Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Oslo, Norway,*Correspondence: Michael A. Tranulis, Department of Basic Sciences and Aquatic Medicine, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences, Campus Adamstuen, Oslo 0033, Norway,
| |
Collapse
|
38
|
Subcellular distribution of the prion protein in sickness and in health. Virus Res 2015; 207:136-45. [PMID: 25683509 DOI: 10.1016/j.virusres.2015.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 11/22/2022]
Abstract
The cellular prion protein (PrP(C)) is an ubiquitously expressed glycoprotein that is most abundant in the central nervous system. It is thought to play a role in many cellular processes, including neuroprotection, but may also contribute to Alzheimer's disease and some cancers. However, it is best known for its central role in the prion diseases, such as Creutzfeldt-Jakob disease (CJD), bovine spongiform encephalopathy (BSE), and scrapie. These protein misfolding diseases can be sporadic, acquired, or genetic and are caused by refolding of endogenous PrP(C) into a beta sheet-rich, pathogenic form, PrP(Sc). Once prions are present in the central nervous system, they increase and spread during a long incubation period that is followed by a relatively short clinical disease phase, ending in death. PrP molecules can be broadly categorized as either 'good' (cellular) PrP(C) or 'bad' (scrapie prion-type) PrP(Sc), but both populations are heterogeneous and different forms of PrP(C) may influence various cellular activities. Both PrP(C) and PrP(Sc) are localized predominantly at the cell surface, with the C-terminus attached to the plasma membrane via a glycosyl-phosphatidylinositol (GPI) anchor and both can exist in cleaved forms. PrP(C) also has cytosolic and transmembrane forms, and PrP(Sc) is known to exist in a variety of conformations and aggregation states. Here, we discuss the roles of different PrP isoforms in sickness and in health, and show the subcellular distributions of several forms of PrP that are particularly relevant for PrP(C) to PrP(Sc) conversion and prion-induced pathology in the hippocampus.
Collapse
|
39
|
Prodromidou K, Papastefanaki F, Sklaviadis T, Matsas R. Functional cross-talk between the cellular prion protein and the neural cell adhesion molecule is critical for neuronal differentiation of neural stem/precursor cells. Stem Cells 2015; 32:1674-87. [PMID: 24497115 DOI: 10.1002/stem.1663] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 12/20/2013] [Accepted: 01/11/2014] [Indexed: 12/15/2022]
Abstract
Cellular prion protein (PrP) is prominently expressed in brain, in differentiated neurons but also in neural stem/precursor cells (NPCs). The misfolding of PrP is a central event in prion diseases, yet the physiological function of PrP is insufficiently understood. Although PrP has been reported to associate with the neural cell adhesion molecule (NCAM), the consequences of concerted PrP-NCAM action in NPC physiology are unknown. Here, we generated NPCs from the subventricular zone (SVZ) of postnatal day 5 wild-type and PrP null (-/-) mice and observed that PrP is essential for proper NPC proliferation and neuronal differentiation. Moreover, we found that PrP is required for the NPC response to NCAM-induced neuronal differentiation. In the absence of PrP, NCAM not only fails to promote neuronal differentiation but also induces an accumulation of doublecortin-positive neuronal progenitors at the proliferation stage. In agreement, we noted an increase in cycling neuronal progenitors in the SVZ of PrP-/- mice compared with PrP+/+ mice, as evidenced by double labeling for the proliferation marker Ki67 and doublecortin as well as by 5-bromo-2'-deoxyuridine incorporation experiments. Additionally, fewer newly born neurons were detected in the rostral migratory stream of PrP-/- mice. Analysis of the migration of SVZ cells in microexplant cultures from wild-type and PrP-/- mice revealed no differences between genotypes or a role for NCAM in this process. Our data demonstrate that PrP plays a critical role in neuronal differentiation of NPCs and suggest that this function is, at least in part, NCAM-dependent.
Collapse
Affiliation(s)
- Kanella Prodromidou
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | | | | | | |
Collapse
|
40
|
Baindur-Hudson S, Edkins AL, Blatch GL. Hsp70/Hsp90 organising protein (hop): beyond interactions with chaperones and prion proteins. Subcell Biochem 2015; 78:69-90. [PMID: 25487016 DOI: 10.1007/978-3-319-11731-7_3] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The Hsp70/Hsp90 organising protein (Hop), also known as stress-inducible protein 1 (STI1), has received considerable attention for diverse cellular functions in both healthy and diseased states. There is extensive evidence that intracellular Hop is a co-chaperone of the major chaperones Hsp70 and Hsp90, playing an important role in the productive folding of Hsp90 client proteins. Consequently, Hop is implicated in a number of key signalling pathways, including aberrant pathways leading to cancer. However, Hop is also secreted and it is now well established that Hop also serves as a receptor for the prion protein, PrP(C). The intracellular and extracellular forms of Hop most likely represent two different isoforms, although the molecular determinants of these divergent functions are yet to be identified. There is also a growing body of research that reports the involvement of Hop in cellular activities that appear independent of either chaperones or PrP(C). While Hop has been shown to have various cellular functions, its biological function remains elusive. However, recent knockout studies in mammals suggest that Hop has an important role in embryonic development. This review provides a critical overview of the latest molecular, cellular and biological research on Hop, critically evaluating its function in healthy systems and how this function is adapted in diseases states.
Collapse
Affiliation(s)
- Swati Baindur-Hudson
- College of Health and Biomedicine, Victoria University, VIC 8001, Melbourne, Australia,
| | | | | |
Collapse
|
41
|
Liebert A, Bicknell B, Adams R. Prion Protein Signaling in the Nervous System—A Review and Perspective. ACTA ACUST UNITED AC 2014. [DOI: 10.4137/sti.s12319] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Prion protein (PrPC) was originally known as the causative agent of transmissible spongiform encephalopathy (TSE) but with recent research, its true function in cells is becoming clearer. It is known to act as a scaffolding protein, binding multiple ligands at the cell membrane and to be involved in signal transduction, passing information from the extracellular matrix (ECM) to the cytoplasm. Its role in the coordination of transmitters at the synapse, glyapse, and gap junction and in short- and long-range neurotrophic signaling gives PrPC a major part in neural transmission and nervous system signaling. It acts to regulate cellular function in multiple targets through its role as a controller of redox status and calcium ion flux. Given the importance of PrPC in cell physiology, this review considers its potential role in disease apart from TSE. The putative functions of PrPC point to involvement in neurodegenerative disease, neuropathic pain, chronic headache, and inflammatory disease including neuroinflammatory disease of the nervous system. Potential targets for the treatment of disease influenced by PrPC are discussed.
Collapse
Affiliation(s)
- Ann Liebert
- Faculty of Health Science, University of Sydney, Australia
| | - Brian Bicknell
- Faculty of Health Science, Australian Catholic University, Australia
| | | |
Collapse
|
42
|
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; 8:266-75. [PMID: 25486050 DOI: 10.4161/pri.32079] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Prion protein, PrP(C), is a glycoprotein that is expressed on the cell surface beginning with the early stages of embryonic stem cell differentiation. Previously, we showed that ectopic expression of PrP(C) in human embryonic stem cells (hESCs) triggered differentiation toward endodermal, mesodermal, and ectodermal lineages, whereas silencing of PrP(C) suppressed differentiation toward ectodermal but not endodermal or mesodermal lineages. Considering that PrP(C) might be involved in controlling the balance between cells of different lineages, the current study was designed to test whether PrP(C) controls differentiation of hESCs into cells of neuron-, oligodendrocyte-, and astrocyte-committed lineages. PrP(C) was silenced in hESCs cultured under three sets of conditions that were previously shown to induce hESCs differentiation into predominantly neuron-, oligodendrocyte-, and astrocyte-committed lineages. We found that silencing of PrP(C) suppressed differentiation toward all three lineages. Similar results were observed in all three protocols, arguing that the effect of PrP(C) was independent of differentiation conditions employed. Moreover, switching PrP(C) expression during a differentiation time course revealed that silencing PrP(C) expression during the very initial stage that corresponds to embryonic bodies has a more significant impact than silencing at later stages of differentiation. The current work illustrates that PrP(C) controls differentiation of hESCs toward neuron-, oligodendrocyte-, and astrocyte-committed lineages and is likely involved at the stage of uncommitted neural progenitor cells rather than lineage-committed neural progenitors.
Collapse
Key Words
- CNTF, ciliary neurotrophic factor
- EBs, embryoid bodies
- EFG, epidermal growth factor
- ESCs, embryonic stem cells
- GFAP, glial fibrillary acidic protein
- GRM, glial restrictive medium
- Lenti-ShPrPC, lentiviral vector expressing short hairpin RNA against PrPC
- Lenti-ShScram, lentiviral vector expressing scrambled shRNA
- Lenti-TetR, lentiviral vector expressing tetracycline repressor
- MEF-CM, mouse embryonic feeder-conditioned medium
- MEFs, mouse embryonic fibroblasts
- NDM, neuronal differentiation medium
- NIM, neural induction medium
- NPM, neural proliferation medium
- Olig1, a marker of oligodendrocyte-committed lineages
- PrPC, normal, cellular isoform of the prion protein
- RA, retinoic acid
- Syn, synapsin I
- TH, tyrosine hydroxylase
- Tet, tetracycline
- TetR, tetracycline repressor
- bFGF, basic fibroblast growth factor
- hES+TetR+ShPrPC, hESCs transfected with Lenti-TetR and Lenti-ShPrPC
- hES+TetR+ShScram, hESCs transfected with Lenti-TetR and Lenti-ShScram
- hESCs, human ESCs
- human embryonic stem cells
- neural progenitor cells
- neuron-committed lineages
- prion protein
- stem cell differentiation
Collapse
Affiliation(s)
- Young Jin Lee
- a Center for Biomedical Engineering and; Technology Department of Anatomy and Neurobiology ; University of Maryland School of Medicine ; Baltimore , MD USA
| | | |
Collapse
|
43
|
Halliez S, Passet B, Martin-Lannerée S, Hernandez-Rapp J, Laude H, Mouillet-Richard S, Vilotte JL, Béringue V. To develop with or without the prion protein. Front Cell Dev Biol 2014; 2:58. [PMID: 25364763 PMCID: PMC4207017 DOI: 10.3389/fcell.2014.00058] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/22/2014] [Indexed: 12/23/2022] Open
Abstract
The deletion of the cellular form of the prion protein (PrPC) in mouse, goat, and cattle has no drastic phenotypic consequence. This stands in apparent contradiction with PrPC quasi-ubiquitous expression and conserved primary and tertiary structures in mammals, and its pivotal role in neurodegenerative diseases such as prion and Alzheimer's diseases. In zebrafish embryos, depletion of PrP ortholog leads to a severe loss-of-function phenotype. This raises the question of a potential role of PrPC in the development of all vertebrates. This view is further supported by the early expression of the PrPC encoding gene (Prnp) in many tissues of the mouse embryo, the transient disruption of a broad number of cellular pathways in early Prnp−/− mouse embryos, and a growing body of evidence for PrPC involvement in the regulation of cell proliferation and differentiation in various types of mammalian stem cells and progenitors. Finally, several studies in both zebrafish embryos and in mammalian cells and tissues in formation support a role for PrPC in cell adhesion, extra-cellular matrix interactions and cytoskeleton. In this review, we summarize and compare the different models used to decipher PrPC functions at early developmental stages during embryo- and organo-genesis and discuss their relevance.
Collapse
Affiliation(s)
- Sophie Halliez
- Institut National de la Recherche Agronomique, U892 Virologie et Immunologie Moléculaires Jouy-en-Josas, France
| | - Bruno Passet
- Institut National de la Recherche Agronomique, UMR1313 Génétique Animale et Biologie Intégrative Jouy-en-Josas, France
| | - Séverine Martin-Lannerée
- Institut National de la Santé et de la Recherche Médicale, UMR-S1124 Paris, France ; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| | - Julia Hernandez-Rapp
- Institut National de la Santé et de la Recherche Médicale, UMR-S1124 Paris, France ; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| | - Hubert Laude
- Institut National de la Recherche Agronomique, U892 Virologie et Immunologie Moléculaires Jouy-en-Josas, France
| | - Sophie Mouillet-Richard
- Institut National de la Santé et de la Recherche Médicale, UMR-S1124 Paris, France ; Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| | - Jean-Luc Vilotte
- Institut National de la Recherche Agronomique, UMR1313 Génétique Animale et Biologie Intégrative Jouy-en-Josas, France
| | - Vincent Béringue
- Institut National de la Recherche Agronomique, U892 Virologie et Immunologie Moléculaires Jouy-en-Josas, France
| |
Collapse
|
44
|
Martin-Lannerée S, Hirsch TZ, Hernandez-Rapp J, Halliez S, Vilotte JL, Launay JM, Mouillet-Richard S. PrP(C) from stem cells to cancer. Front Cell Dev Biol 2014; 2:55. [PMID: 25364760 PMCID: PMC4207012 DOI: 10.3389/fcell.2014.00055] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/11/2014] [Indexed: 12/23/2022] Open
Abstract
The cellular prion protein PrP(C) was initially discovered as the normal counterpart of the pathological scrapie prion protein PrP(Sc), the main component of the infectious agent of Transmissible Spongiform Encephalopathies. While clues as to the physiological function of this ubiquitous protein were greatly anticipated from the development of knockout animals, PrP-null mice turned out to be viable and to develop without major phenotypic abnormalities. Notwithstanding, the discovery that hematopoietic stem cells from PrP-null mice have impaired long-term repopulating potential has set the stage for investigating into the role of PrP(C) in stem cell biology. A wealth of data have now exemplified that PrP(C) is expressed in distinct types of stem cells and regulates their self-renewal as well as their differentiation potential. A role for PrP(C) in the fate restriction of embryonic stem cells has further been proposed. Paralleling these observations, an overexpression of PrP(C) has been documented in various types of tumors. In line with the contribution of PrP(C) to stemness and to the proliferation of cancer cells, PrP(C) was recently found to be enriched in subpopulations of tumor-initiating cells. In the present review, we summarize the current knowledge of the role played by PrP(C) in stem cell biology and discuss how the subversion of its function may contribute to cancer progression.
Collapse
Affiliation(s)
- Séverine Martin-Lannerée
- Toxicology, Pharmacology and Cellular Signaling, INSERM UMR-S1124 Paris, France ; Toxicology, Pharmacology and Cellular Signaling, Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| | - Théo Z Hirsch
- Toxicology, Pharmacology and Cellular Signaling, INSERM UMR-S1124 Paris, France ; Toxicology, Pharmacology and Cellular Signaling, Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| | - Julia Hernandez-Rapp
- Toxicology, Pharmacology and Cellular Signaling, INSERM UMR-S1124 Paris, France ; Toxicology, Pharmacology and Cellular Signaling, Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France ; Université Paris Sud 11, ED419 Biosigne Orsay, France
| | - Sophie Halliez
- U892 Virologie et Immunologie Moléculaires, INRA Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- UMR1313 Génétique Animale et Biologie Intégrative, INRA Jouy-en-Josas, France
| | - Jean-Marie Launay
- AP-HP Service de Biochimie, Fondation FondaMental, INSERM U942 Hôpital Lariboisière Paris, France ; Pharma Research Department, F. Hoffmann-La-Roche Ltd. Basel, Switzerland
| | - Sophie Mouillet-Richard
- Toxicology, Pharmacology and Cellular Signaling, INSERM UMR-S1124 Paris, France ; Toxicology, Pharmacology and Cellular Signaling, Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| |
Collapse
|
45
|
Zhang S, Wu D, Wang J, Wang Y, Wang G, Yang M, Yang X. Stress protein expression in early phase spinal cord ischemia/reperfusion injury. Neural Regen Res 2014; 8:2225-35. [PMID: 25206532 PMCID: PMC4146036 DOI: 10.3969/j.issn.1673-5374.2013.24.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/17/2013] [Indexed: 12/05/2022] Open
Abstract
Spinal cord ischemia/reperfusion injury is a stress injury to the spinal cord. Our previous studies using differential proteomics identified 21 differentially expressed proteins (n > 2) in rabbits with spinal cord ischemia/reperfusion injury. Of these proteins, stress-related proteins included protein disulfide isomerase A3, stress-induced-phosphoprotein 1 and heat shock cognate protein 70. In this study, we established New Zealand rabbit models of spinal cord ischemia/reperfusion injury by abdominal aorta occlusion. Results demonstrated that hind limb function initially improved after spinal cord ischemia/reperfusion injury, but then deteriorated. The pathological morphology of the spinal cord became aggravated, but lessened 24 hours after reperfusion. However, the numbers of motor neurons and interneurons in the spinal cord gradually decreased. The expression of protein disulfide isomerase A3, stress-induced-phosphoprotein 1 and heat shock cognate protein 70 was induced by ischemia/reperfusion injury. The expression of these proteins increased within 12 hours after reperfusion, and then decreased, reached a minimum at 24 hours, but subsequently increased again to similar levels seen at 6–12 hours, showing a characterization of induction-inhibition-induction. These three proteins were expressed only in cytoplasm but not in the nuclei. Moreover, the expression was higher in interneurons than in motor neurons, and the survival rate of interneurons was greater than that of motor neurons. It is assumed that the expression of stress-related proteins exhibited a protective effect on neurons.
Collapse
Affiliation(s)
- Shanyong Zhang
- Team of Spine and Spinal Cord, Department of Orthopedics, China-Japan Friendship Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Dankai Wu
- Team of Skeletal Trauma, Department of Orthopedics, the Second Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Jincheng Wang
- Team of Skeletal Trauma, Department of Orthopedics, the Second Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Yongming Wang
- Team of Spine and Spinal Cord, Department of Orthopedics, China-Japan Friendship Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Guoxiang Wang
- Team of Spine and Spinal Cord, Department of Orthopedics, China-Japan Friendship Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Maoguang Yang
- Team of Spine and Spinal Cord, Department of Orthopedics, China-Japan Friendship Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Xiaoyu Yang
- Team of Spine and Spinal Cord, Department of Orthopedics, China-Japan Friendship Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
46
|
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: 3.1] [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
|
47
|
STI1 antagonizes cytoskeleton collapse mediated by small GTPase Rnd1 and regulates neurite growth. Exp Cell Res 2014; 324:84-91. [PMID: 24690281 DOI: 10.1016/j.yexcr.2014.03.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 03/19/2014] [Accepted: 03/22/2014] [Indexed: 01/10/2023]
Abstract
Rnd proteins comprise a branch of the Rho family of small GTP-binding proteins, which have been implicated in rearrangements of the actin cytoskeleton and microtubule dynamics. Particularly in the nervous system, Rnd family proteins regulate neurite formation, dendrite development and axonal branching. A secreted form of the co-chaperone Stress-Inducible Protein 1 (STI1) has been described as a prion protein partner that is involved in several processes of the nervous system, such as neurite outgrowth, neuroprotection, astrocyte development, and the self-renewal of neural progenitor cells. We show that cytoplasmic STI1 directly interacts with the GTPase Rnd1. This interaction is specific for the Rnd1 member of the Rnd family. In the COS collapse assay, overexpression of STI1 prevents Rnd1-plexin-A1-mediated cytoskeleton retraction. In PC-12 cells, overexpression of STI1 enhances neurite outgrowth in cellular processes initially established by Rnd1. Therefore, we propose that STI1 participates in Rnd1-induced signal transduction pathways that are involved in the dynamics of the actin cytoskeleton.
Collapse
|
48
|
Prion protein misfolding, strains, and neurotoxicity: an update from studies on Mammalian prions. Int J Cell Biol 2013; 2013:910314. [PMID: 24454379 PMCID: PMC3884631 DOI: 10.1155/2013/910314] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 11/10/2013] [Accepted: 11/11/2013] [Indexed: 11/17/2022] Open
Abstract
Prion diseases, also known as transmissible spongiform encephalopathies (TSEs), are a group of fatal neurodegenerative disorders affecting humans and other mammalian species. The central event in TSE pathogenesis is the conformational conversion of the cellular prion protein, PrPC, into the aggregate, β-sheet rich, amyloidogenic form, PrPSc. Increasing evidence indicates that distinct PrPSc conformers, forming distinct ordered aggregates, can encipher the phenotypic TSE variants related to prion strains. Prion strains are TSE isolates that, after inoculation into syngenic hosts, cause disease with distinct characteristics, such as incubation period, pattern of PrPSc distribution, and regional severity of histopathological changes in the brain. In analogy with other amyloid forming proteins, PrPSc toxicity is thought to derive from the existence of various intermediate structures prior to the amyloid fiber formation and/or their specific interaction with membranes. The latter appears particularly relevant for the pathogenesis of TSEs associated with GPI-anchored PrPSc, which involves major cellular membrane distortions in neurons. In this review, we update the current knowledge on the molecular mechanisms underlying three fundamental aspects of the basic biology of prions such as the putative mechanism of prion protein conversion to the pathogenic form PrPSc and its propagation, the molecular basis of prion strains, and the mechanism of induced neurotoxicity by PrPSc aggregates.
Collapse
|
49
|
Llorens F, Ferrer I, del Río JA. Gene expression resulting from PrPC ablation and PrPC overexpression in murine and cellular models. Mol Neurobiol 2013; 49:413-23. [PMID: 23949728 DOI: 10.1007/s12035-013-8529-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/05/2013] [Indexed: 02/07/2023]
Abstract
The cellular prion protein (PrP(C)) plays a key role in prion diseases when it converts to the pathogenic form scrapie prion protein. Increasing knowledge of its participation in prion infection contrasts with the elusive and controversial data regarding its physiological role probably related to its pleiotropy, cell-specific functions, and cellular-specific milieu. Multiple approaches have been made to the increasing understanding of the molecular mechanisms and cellular functions modulated by PrP(C) at the transcriptomic and proteomic levels. Gene expression analyses have been made in several mouse and cellular models with regulated expression of PrP(C) resulting in PrP(C) ablation or PrP(C) overexpression. These analyses support previous functional data and have yielded clues about new potential functions. However, experiments on animal models have shown moderate and varied results which are difficult to interpret. Moreover, studies in cell cultures correlate little with in vivo counterparts. Yet, both animal and cell models have provided some insights on how to proceed in the future by using more refined methods and selected functional experiments.
Collapse
Affiliation(s)
- Franc Llorens
- Institute of Neuropathology, University Hospital Bellvitge-Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain,
| | | | | |
Collapse
|
50
|
Relaño-Ginès A, Gabelle A, Hamela C, Belondrade M, Casanova D, Mourton-Gilles C, Lehmann S, Crozet C. Prion replication occurs in endogenous adult neural stem cells and alters their neuronal fate: involvement of endogenous neural stem cells in prion diseases. PLoS Pathog 2013; 9:e1003485. [PMID: 23935493 PMCID: PMC3731238 DOI: 10.1371/journal.ppat.1003485] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 05/24/2013] [Indexed: 11/18/2022] Open
Abstract
Prion diseases are irreversible progressive neurodegenerative diseases, leading to severe incapacity and death. They are characterized in the brain by prion amyloid deposits, vacuolisation, astrocytosis, neuronal degeneration, and by cognitive, behavioural and physical impairments. There is no treatment for these disorders and stem cell therapy therefore represents an interesting new approach. Gains could not only result from the cell transplantation, but also from the stimulation of endogenous neural stem cells (NSC) or by the combination of both approaches. However, the development of such strategies requires a detailed knowledge of the pathology, particularly concerning the status of the adult neurogenesis and endogenous NSC during the development of the disease. During the past decade, several studies have consistently shown that NSC reside in the adult mammalian central nervous system (CNS) and that adult neurogenesis occurs throughout the adulthood in the subventricular zone of the lateral ventricle or the Dentate Gyrus of the hippocampus. Adult NSC are believed to constitute a reservoir for neuronal replacement during normal cell turnover or after brain injury. However, the activation of this system does not fully compensate the neuronal loss that occurs during neurodegenerative diseases and could even contribute to the disease progression. We investigated here the status of these cells during the development of prion disorders. We were able to show that NSC accumulate and replicate prions. Importantly, this resulted in the alteration of their neuronal fate which then represents a new pathologic event that might underlie the rapid progression of the disease.
Collapse
Affiliation(s)
| | - Audrey Gabelle
- Institut de Génétique Humaine, CNRS-UPR 1142, Montpellier, France
- Institut de Recherche en Biothérapie (IRB), Physiopathologie, Diagnostic et Thérapie Cellulaire des Affections Neurodégénératives - INSERM-UM1 U1040, CHU de Montpellier, Université Montpellier 1, Montpellier, France
| | - Claire Hamela
- Institut de Génétique Humaine, CNRS-UPR 1142, Montpellier, France
| | | | | | | | - Sylvain Lehmann
- Institut de Génétique Humaine, CNRS-UPR 1142, Montpellier, France
- Institut de Recherche en Biothérapie (IRB), Physiopathologie, Diagnostic et Thérapie Cellulaire des Affections Neurodégénératives - INSERM-UM1 U1040, CHU de Montpellier, Université Montpellier 1, Montpellier, France
- * E-mail: (SL); (CC)
| | - Carole Crozet
- Institut de Génétique Humaine, CNRS-UPR 1142, Montpellier, France
- Institut de Recherche en Biothérapie (IRB), Physiopathologie, Diagnostic et Thérapie Cellulaire des Affections Neurodégénératives - INSERM-UM1 U1040, CHU de Montpellier, Université Montpellier 1, Montpellier, France
- * E-mail: (SL); (CC)
| |
Collapse
|