1
|
Huntingtin and Other Neurodegeneration-Associated Proteins in the Development of Intracellular Pathologies: Potential Target Search for Therapeutic Intervention. Int J Mol Sci 2022; 23:ijms232415533. [PMID: 36555175 PMCID: PMC9779313 DOI: 10.3390/ijms232415533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are currently incurable. Numerous experimental data accumulated over the past fifty years have brought us closer to understanding the molecular and cell mechanisms responsible for their development. However, these data are not enough for a complete understanding of the genesis of these diseases, nor to suggest treatment methods. It turns out that many cellular pathologies developing during neurodegeneration coincide from disease to disease. These observations give hope to finding a common intracellular target(s) and to offering a universal method of treatment. In this review, we attempt to analyze data on similar cellular disorders among neurodegenerative diseases in general, and polyglutamine neurodegenerative diseases in particular, focusing on the interaction of various proteins involved in the development of neurodegenerative diseases with various cellular organelles. The main purposes of this review are: (1) to outline the spectrum of common intracellular pathologies and to answer the question of whether it is possible to find potential universal target(s) for therapeutic intervention; (2) to identify specific intracellular pathologies and to speculate about a possible general approach for their treatment.
Collapse
|
2
|
Grimaldi I, Leser FS, Janeiro JM, da Rosa BG, Campanelli AC, Romão L, Lima FRS. The multiple functions of PrP C in physiological, cancer, and neurodegenerative contexts. J Mol Med (Berl) 2022; 100:1405-1425. [PMID: 36056255 DOI: 10.1007/s00109-022-02245-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022]
Abstract
Cellular prion protein (PrPC) is a highly conserved glycoprotein, present both anchored in the cell membrane and soluble in the extracellular medium. It has a diversity of ligands and is variably expressed in numerous tissues and cell subtypes, most notably in the central nervous system (CNS). Its importance has been brought to light over the years both under physiological conditions, such as embryogenesis and immune system homeostasis, and in pathologies, such as cancer and neurodegenerative diseases. During development, PrPC plays an important role in CNS, participating in axonal growth and guidance and differentiation of glial cells, but also in other organs such as the heart, lung, and digestive system. In diseases, PrPC has been related to several types of tumors, modulating cancer stem cells, enhancing malignant properties, and inducing drug resistance. Also, in non-neoplastic diseases, such as Alzheimer's and Parkinson's diseases, PrPC seems to alter the dynamics of neurotoxic aggregate formation and, consequently, the progression of the disease. In this review, we explore in detail the multiple functions of this protein, which proved to be relevant for understanding the dynamics of organism homeostasis, as well as a promising target in the treatment of both neoplastic and degenerative diseases.
Collapse
Affiliation(s)
- Izabella Grimaldi
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Felipe Saceanu Leser
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José Marcos Janeiro
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bárbara Gomes da Rosa
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Clara Campanelli
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luciana Romão
- Cell Morphogenesis Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Flavia Regina Souza Lima
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
3
|
Loh D, Reiter RJ. Melatonin: Regulation of Prion Protein Phase Separation in Cancer Multidrug Resistance. Molecules 2022; 27:705. [PMID: 35163973 PMCID: PMC8839844 DOI: 10.3390/molecules27030705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
The unique ability to adapt and thrive in inhospitable, stressful tumor microenvironments (TME) also renders cancer cells resistant to traditional chemotherapeutic treatments and/or novel pharmaceuticals. Cancer cells exhibit extensive metabolic alterations involving hypoxia, accelerated glycolysis, oxidative stress, and increased extracellular ATP that may activate ancient, conserved prion adaptive response strategies that exacerbate multidrug resistance (MDR) by exploiting cellular stress to increase cancer metastatic potential and stemness, balance proliferation and differentiation, and amplify resistance to apoptosis. The regulation of prions in MDR is further complicated by important, putative physiological functions of ligand-binding and signal transduction. Melatonin is capable of both enhancing physiological functions and inhibiting oncogenic properties of prion proteins. Through regulation of phase separation of the prion N-terminal domain which targets and interacts with lipid rafts, melatonin may prevent conformational changes that can result in aggregation and/or conversion to pathological, infectious isoforms. As a cancer therapy adjuvant, melatonin could modulate TME oxidative stress levels and hypoxia, reverse pH gradient changes, reduce lipid peroxidation, and protect lipid raft compositions to suppress prion-mediated, non-Mendelian, heritable, but often reversible epigenetic adaptations that facilitate cancer heterogeneity, stemness, metastasis, and drug resistance. This review examines some of the mechanisms that may balance physiological and pathological effects of prions and prion-like proteins achieved through the synergistic use of melatonin to ameliorate MDR, which remains a challenge in cancer treatment.
Collapse
Affiliation(s)
- Doris Loh
- Independent Researcher, Marble Falls, TX 78654, USA
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
4
|
Li R, Ren M, Yu Y. Anti-PrP monoclonal antibody as a novel treatment for neurogenesis in mouse model of Alzheimer's disease. Brain Behav 2021; 11:e2365. [PMID: 34672433 PMCID: PMC8613428 DOI: 10.1002/brb3.2365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 08/25/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common degenerative disease characterized by cognitive impairment, memory decline, and language disorder for which there is no effective treatment. Neurogenesis has been indicated in AD and may play an important role in the pathogenesis of AD. Targeting this pathway is a new idea for the treatment of the disease. A recent study reveals that the cellular prion protein (PrP), a receptor for Aβ oligomers, regulates neurogenesis, and its elevated expression is related to cell differentiation. The aim of the present study was to investigate the neuroprotective effects of 6D11 (PrP monoclonal antibody) via neurogenesis promotion in APP/PS1 transgenic mice and Aβ-induced cell model of AD. METHODS In the present study, 9-month-old male APP/PS1 mice were injected with 6D11. Then, the Morris water maze was used to examine the spatial learning and memory abilities of the mice in both groups, and immunostained was used to assess the level of Aβ, neurogenesis, and neural stem cells (NSCs) differentiation. RESULTS 6D11 attenuated cognitive deficits in APP/PS1 transgenic mice, which was accompanied by a decrease of the deposition of Aβ. In addition, 6D11 treatment promoted differentiation of the existing hippocampal cells to neurons. CONCLUSIONS Our findings confirmed that 6D11 has a therapeutic effect in APP/PS1 transgenic AD mouse model and Aβ-induced AD cell model, and the effect exerted via increase of neurogenesis and cell differentiation by transduction of Aβ peptide signal.
Collapse
Affiliation(s)
- Ruolin Li
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, China
| | - Ming Ren
- Department of neurology, Shanghai blue cross brain hospital, 2880 Qixin Road, Minhang District, Shanghai, 201101, China
| | - Yingxin Yu
- Department of Neurology, Chinese PLA General Hospital, 28th Fuxing Road, Haidian district, Beijing, 100048, China
| |
Collapse
|
5
|
Fremuntova Z, Mosko T, Soukup J, Kucerova J, Kostelanska M, Hanusova ZB, Filipova M, Cervenakova L, Holada K. Changes in cellular prion protein expression, processing and localisation during differentiation of the neuronal cell line CAD 5. Biol Cell 2019; 112:1-21. [PMID: 31736091 DOI: 10.1111/boc.201900045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND INFORMATION Cellular prion protein (PrPC ) is infamous for its role in prion diseases. The physiological function of PrPC remains enigmatic, but several studies point to its involvement in cell differentiation processes. To test this possibility, we monitored PrPC changes during the differentiation of prion-susceptible CAD 5 cells, and then we analysed the effect of PrPC ablation on the differentiation process. RESULTS Neuronal CAD 5 cells differentiate within 5 days of serum withdrawal, with the majority of the cells developing long neurites. This process is accompanied by an up to sixfold increase in PrPC expression and enhanced N-terminal β-cleavage of the protein, which suggests a role for the PrPC in the differentiation process. Moreover, the majority of PrPC in differentiated cells is inside the cell, and a large proportion of the protein does not associate with membrane lipid rafts. In contrast, PrPC in proliferating cells is found mostly on the cytoplasmic membrane and is predominantly associated with lipid rafts. To determine the importance of PrPC in cell differentiation, a CAD 5 PrP-/- cell line with ablated PrPC expression was created using the CRISPR/Cas9 system. We observed no considerable difference in morphology, proliferation rate or expression of molecular markers between CAD 5 and CAD 5 PrP-/- cells during the differentiation initiated by serum withdrawal. CONCLUSIONS PrPC characteristics, such as cell localisation, level of expression and posttranslational modifications, change during CAD 5 cell differentiation, but PrPC ablation does not change the course of the differentiation process. SIGNIFICANCE Ablation of PrPC expression does not affect CAD 5 cell differentiation, although we observed many intriguing changes in PrPC features during the process. Our study does not support the concept that PrPC is important for neuronal cell differentiation, at least in simple in vitro conditions.
Collapse
Affiliation(s)
- Zuzana Fremuntova
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tibor Mosko
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jakub Soukup
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.,Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Johanka Kucerova
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Marie Kostelanska
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Zdenka Backovska Hanusova
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Marcela Filipova
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | | | - Karel Holada
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
6
|
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
|
7
|
Jeong JK, Lee JH, Kim SW, Hong JM, Seol JW, Park SY. Cellular prion protein regulates the differentiation and function of adipocytes through autophagy flux. Mol Cell Endocrinol 2019; 481:84-94. [PMID: 30513342 DOI: 10.1016/j.mce.2018.11.013] [Citation(s) in RCA: 5] [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: 08/24/2018] [Revised: 11/06/2018] [Accepted: 11/23/2018] [Indexed: 02/06/2023]
Abstract
The role of autophagy modulation in adipogenic differentiation and the possible autophagy modulators targeting adipogenesis remain unclear. In this study, we investigated whether normal cellular prion protein (PrP<C>) is involved in the modulation of autophagy and affects adipogenic differentiation in vivo and in vitro. Surprisingly, autophagy flux signals were activated in the adipose tissue of prion protein-deficient mice and PrP<C>-deleted 3T3-L1 adipocytes. The activation of autophagy flux mediated by PrP<C> deletion was confirmed in the adipose tissue via transmission electron microscopy. Adipocyte differentiation factors were highly induced in prion protein-deficient adipose tissue and 3T3-L1 adipocytes. In addition, deletion of prion protein significantly increased visceral fat volume, body fat weight, adipocyte cell size, and body weight gain in Prnp-knockout mice and increased lipid accumulation in PrP<C> siRNA-transfected 3T3-L1 cells. However, the overexpression of prion protein using adenovirus inhibited the autophagic flux signals, lipid accumulation, and the PPAR-γ and C/EBP-α mRNA and protein expression levels in comparison to those in the control cells. Our results demonstrated that deletion of normal prion protein accelerated adipogenic differentiation and lipid accumulation mediated via autophagy flux activation.
Collapse
Affiliation(s)
- Jae-Kyo Jeong
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Ju-Hee Lee
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Sung-Wook Kim
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Jeong-Min Hong
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea.
| |
Collapse
|
8
|
Lee JH, Yun CW, Han YS, Kim S, Jeong D, Kwon HY, Kim H, Baek MJ, Lee SH. Melatonin and 5-fluorouracil co-suppress colon cancer stem cells by regulating cellular prion protein-Oct4 axis. J Pineal Res 2018; 65:e12519. [PMID: 30091203 DOI: 10.1111/jpi.12519] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/12/2018] [Accepted: 07/31/2018] [Indexed: 12/21/2022]
Abstract
Melatonin suppresses tumor development. However, the exact relationship between melatonin and cancer stem cells (CSCs) is poorly understood. This study found that melatonin inhibits colon CSCs by regulating the PrPC -Oct4 axis. In specimens from patients with colorectal cancer, the expressions of cellular prion protein (PrPC ) and Oct4 were significantly correlated with metastasis and tumor stages. Co-treatment with 5-fluorouracil (5-FU) and melatonin inhibited the stem cell markers Oct4, Nanog, Sox2, and ALDH1A1 by downregulating PrPC . In this way, tumor growth, proliferation, and tumor-mediated angiogenesis were suppressed. In colorectal CSCs, PRNP overexpression protects Oct4 against inhibition by 5-FU and melatonin. In contrast, Nanog, Sox2, and ALDH1A1 have no such protection. These results indicate that PrPC directly regulates Oct4, whereas it indirectly regulates Nanog, Sox2, and ALDH1A1. Taken together, our findings suggest that co-treatment with anticancer drug and melatonin is a potential therapy for colorectal cancer. Furthermore, PrPC maintains cancer stemness during tumor progression. Therefore, targeting the PrPC -Oct4 axis may prove instrumental in colorectal cancer therapy.
Collapse
Affiliation(s)
- Jun Hee Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Chul Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - SangMin Kim
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Dongjun Jeong
- Department of Pathology, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Hyog Young Kwon
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Hyeongjoo Kim
- Soonchunhyang Medical Science Research Institute, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Moo-Jun Baek
- Department of Surgery, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
9
|
Collins SJ, Tumpach C, Groveman BR, Drew SC, Haigh CL. Prion protein cleavage fragments regulate adult neural stem cell quiescence through redox modulation of mitochondrial fission and SOD2 expression. Cell Mol Life Sci 2018; 75:3231-3249. [PMID: 29574582 PMCID: PMC6063333 DOI: 10.1007/s00018-018-2790-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 02/27/2018] [Accepted: 03/05/2018] [Indexed: 01/06/2023]
Abstract
Neurogenesis continues in the post-developmental brain throughout life. The ability to stimulate the production of new neurones requires both quiescent and actively proliferating pools of neural stem cells (NSCs). Actively proliferating NSCs ensure that neurogenic demand can be met, whilst the quiescent pool makes certain NSC reserves do not become depleted. The processes preserving the NSC quiescent pool are only just beginning to be defined. Herein, we identify a switch between NSC proliferation and quiescence through changing intracellular redox signalling. We show that N-terminal post-translational cleavage products of the prion protein (PrP) induce a quiescent state, halting NSC cellular growth, migration, and neurite outgrowth. Quiescence is initiated by the PrP cleavage products through reducing intracellular levels of reactive oxygen species. First, inhibition of redox signalling results in increased mitochondrial fission, which rapidly signals quiescence. Thereafter, quiescence is maintained through downstream increases in the expression and activity of superoxide dismutase-2 that reduces mitochondrial superoxide. We further observe that PrP is predominantly cleaved in quiescent NSCs indicating a homeostatic role for this cascade. Our findings provide new insight into the regulation of NSC quiescence, which potentially could influence brain health throughout adult life.
Collapse
Affiliation(s)
- Steven J Collins
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Carolin Tumpach
- Doherty Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Bradley R Groveman
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, 59840, USA
| | - Simon C Drew
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Cathryn L Haigh
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Melbourne, VIC, 3010, Australia.
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, 59840, USA.
| |
Collapse
|
10
|
Martellucci S, Manganelli V, Santacroce C, Santilli F, Piccoli L, Sorice M, Mattei V. Role of Prion protein-EGFR multimolecular complex during neuronal differentiation of human dental pulp-derived stem cells. Prion 2018; 12:117-126. [PMID: 29644924 DOI: 10.1080/19336896.2018.1463797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cellular prion protein (PrPC) is expressed in a wide variety of stem cells in which regulates their self-renewal as well as differentiation potential. In this study we investigated the presence of PrPC in human dental pulp-derived stem cells (hDPSCs) and its role in neuronal differentiation process. We show that hDPSCs expresses early PrPC at low concentration and its expression increases after two weeks of treatment with EGF/bFGF. Then, we analyzed the association of PrPC with gangliosides and EGF receptor (EGF-R) during neuronal differentiation process. PrPC associates constitutively with GM2 in control hDPSCs and with GD3 only after neuronal differentiation. Otherwise, EGF-R associates weakly in control hDPSCs and more markedly after neuronal differentiation. To analyze the functional role of PrPC in the signal pathway mediated by EGF/EGF-R, a siRNA PrP was applied to ablate PrPC and its function. The treatment with siRNA PrP significantly prevented Akt and ERK1/2 phosphorylation induced by EGF. Moreover, siRNA PrP treatment significantly prevented neuronal-specific antigens expression induced by EGF/bFGF, indicating that cellular prion protein is essential for EGF/bFGF-induced hDPSCs differentiation. These results suggest that PrPC interact with EGF-R within lipid rafts, playing a role in the multimolecular signaling complexes involved in hDPSCs neuronal differentiation.
Collapse
Affiliation(s)
- Stefano Martellucci
- a Laboratory of Experimental Medicine and Environmental Pathology - Rieti University Hub "Sabina Universitas" , Via Angelo Maria Ricci 35/A, Rieti , Italy.,b Department of Experimental Medicine - "Sapienza" University , Viale Regina Elena 324, Rome , Italy
| | - Valeria Manganelli
- b Department of Experimental Medicine - "Sapienza" University , Viale Regina Elena 324, Rome , Italy
| | - Costantino Santacroce
- a Laboratory of Experimental Medicine and Environmental Pathology - Rieti University Hub "Sabina Universitas" , Via Angelo Maria Ricci 35/A, Rieti , Italy
| | - Francesca Santilli
- a Laboratory of Experimental Medicine and Environmental Pathology - Rieti University Hub "Sabina Universitas" , Via Angelo Maria Ricci 35/A, Rieti , Italy
| | - Luca Piccoli
- c Department of Science Dentistry and Maxillofacial - "Sapienza" University , Viale Regina Elena 287/A, Rome , Italy
| | - Maurizio Sorice
- b Department of Experimental Medicine - "Sapienza" University , Viale Regina Elena 324, Rome , Italy
| | - Vincenzo Mattei
- a Laboratory of Experimental Medicine and Environmental Pathology - Rieti University Hub "Sabina Universitas" , Via Angelo Maria Ricci 35/A, Rieti , Italy.,b Department of Experimental Medicine - "Sapienza" University , Viale Regina Elena 324, Rome , Italy
| |
Collapse
|
11
|
Role of hypoxia‑mediated cellular prion protein functional change in stem cells and potential application in angiogenesis (Review). Mol Med Rep 2017; 16:5747-5751. [PMID: 28901450 PMCID: PMC5865755 DOI: 10.3892/mmr.2017.7387] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 06/19/2017] [Indexed: 12/22/2022] Open
Abstract
Cellular prion protein (PrPC) can replace other pivotal molecules due to its interaction with several partners in performing a variety of important biological functions that may differ between embryonic and mature stem cells. Recent studies have revealed major advances in elucidating the putative role of PrPC in the regulation of stem cells and its application in stem cell therapy. What is special about PrPC is that its expression may be regulated by hypoxia-inducible factor (HIF)-1α, which is the transcriptional factor of cellular response to hypoxia. Hypoxic conditions have been known to drive cellular responses that can enhance cell survival, differentiation and angiogenesis through adaptive processes. Our group recently reported hypoxia-enhanced vascular repair of endothelial colony-forming cells on ischemic injury. Hypoxia-induced AKT/signal transducer and activator of transcription 3 phosphorylation eventually increases neovasculogenesis. In stem cell biology, hypoxia promotes the expression of growth factors. According to other studies, aspects of tissue regeneration and cell function are influenced by hypoxia, which serves an essential role in stem cell HIF-1α signaling. All these data suggest the possibility that hypoxia-mediated PrPC serves an important role in angiogenesis. Therefore, the present review summarizes the characteristics of PrPC, which is produced by HIF-1α in hypoxia, as it relates to angiogenesis.
Collapse
|
12
|
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
|
13
|
Wan X, Yang R, Liu G, Zhu M, Zhang T, Liu L, Wu R. Downregulation of cellular prion protein inhibited the proliferation and invasion and induced apoptosis of Marek's disease virus-transformed avian T cells. J Vet Sci 2017; 17:171-8. [PMID: 26243599 PMCID: PMC4921665 DOI: 10.4142/jvs.2016.17.2.171] [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: 02/03/2015] [Revised: 04/30/2015] [Accepted: 07/03/2015] [Indexed: 11/20/2022] Open
Abstract
Cellular prion protein (PrP(C)) is ubiquitously expressed in the cytomembrane of a considerable number of eukaryotic cells. Although several studies have investigated the functions of PrP(C) in cell proliferation, cell apoptosis, and tumorigenesis of mammals, the correlated functions of chicken PrP(C) (chPrP(C)) remain unknown. In this study, stable chPrP(C)-downregulated Marek's disease (MD) virus-transformed avian T cells (MSB1-SiRNA-3) were established by introducing short interfering RNA (SiRNA) targeting chicken prion protein genes. We found that downregulation of chPrP(C) inhibits proliferation, invasion, and migration, and induces G1 cell cycle phase arrest and apoptosis of MSB1-SiRNA-3 cells compared with Marek's disease virus-transformed avian T cells (MSB1) and negative control cells. To the best of our knowledge, the present study provides the first evidence supporting the positive correlation between the expression level of chPrP(C) and the proliferation, migration, and invasion ability of MSB1 cells, but appears to protect MSB1 cells from apoptosis, which suggests it functions in the formation and development of MD tumors. This evidence may contribute to future research into the specific molecular mechanisms of chPrP(C) in the formation and development of MD tumors.
Collapse
Affiliation(s)
- Xuerui Wan
- Department of Preventive Veterinary, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Runxia Yang
- Department of Preventive Veterinary, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Guilin Liu
- Department of Preventive Veterinary, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Manling Zhu
- Department of Preventive Veterinary, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Tianliang Zhang
- Department of Preventive Veterinary, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Lei Liu
- Department of Preventive Veterinary, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Run Wu
- Department of Preventive Veterinary, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
14
|
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
|
15
|
Dalai W, Matsuo E, Takeyama N, Kawano J, Saeki K. Increased expression of prion protein gene is accompanied by demethylation of CpG sites in a mouse embryonal carcinoma cell line, P19C6. J Vet Med Sci 2017; 79:644-648. [PMID: 28132962 PMCID: PMC5383191 DOI: 10.1292/jvms.16-0554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Elucidation of the processes regulating the prion protein gene (Prnp) is an important key to understanding the development of prion disorders.
In this study, we explored the involvement of DNA methylation in Prnp transcriptional regulation during neuronal differentiation of embryonic
carcinoma P19C6 cells. When P19C6 cells were differentiated into neuronal cells, the expression of Prnp was markedly increased, while CpG
methylation was significantly demethylated at the nucleotide region between −599 and −238 from the transcription start site. In addition, when P19C6 cells were
applied in a DNA methyltransferase inhibitor, RG108, Prnp transcripts were also significantly increased in relation to the decreased
methylation statuses. These findings helped to elucidate the DNA methylation-mediated regulation of Prnp expression during neuronal
differentiation.
Collapse
Affiliation(s)
- Wuyun Dalai
- Laboratory of Microbiology and Immunology, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan
| | | | | | | | | |
Collapse
|
16
|
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
|
17
|
ONODERA T. Dual role of cellular prion protein in normal host and Alzheimer's disease. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:155-173. [PMID: 28413194 PMCID: PMC5489426 DOI: 10.2183/pjab.93.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/26/2017] [Indexed: 06/07/2023]
Abstract
Using PrPC-knockout cell lines, it has been shown that the inhibition of apoptosis through STI1 is mediated by PrPC-dependent SOD activation. Antioxidant PrPC may contribute to suppression of inflammasome activation. PrPC is functionally involved in copper metabolism, signal transduction, neuroprotection, and cell maturation. Recently several reports have shown that PrPC participates in trans-membrane signaling processes associated with hematopoietic stem cell replication and neuronal differentiation. In another role, PrPC also tends to function as a neurotoxic protein. Aβ oligomer, which is associated with neurodegeneration in Alzheimer's disease (AD), has also been reported to act as a ligand of PrPC. However, the physiological role of PrPC as an Aβ42-binding protein is not clear. Actually, PrPC is critical in Aβ42-mediated autophagy in neurons. PrPC shows a beneficial role in lipid rafts to promote autophagy. Further search for PrPC-interaction molecules using Prnp-/- mice and various types of Prnp-/- cell lines under various conditions may elucidate other important PrPC important functions.
Collapse
Affiliation(s)
- Takashi ONODERA
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
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
|
19
|
Sinclair KD, Rutherford KMD, Wallace JM, Brameld JM, Stöger R, Alberio R, Sweetman D, Gardner DS, Perry VEA, Adam CL, Ashworth CJ, Robinson JE, Dwyer CM. Epigenetics and developmental programming of welfare and production traits in farm animals. Reprod Fertil Dev 2016; 28:RD16102. [PMID: 27439952 DOI: 10.1071/rd16102] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/06/2016] [Indexed: 12/11/2022] Open
Abstract
The concept that postnatal health and development can be influenced by events that occur in utero originated from epidemiological studies in humans supported by numerous mechanistic (including epigenetic) studies in a variety of model species. Referred to as the 'developmental origins of health and disease' or 'DOHaD' hypothesis, the primary focus of large-animal studies until quite recently had been biomedical. Attention has since turned towards traits of commercial importance in farm animals. Herein we review the evidence that prenatal risk factors, including suboptimal parental nutrition, gestational stress, exposure to environmental chemicals and advanced breeding technologies, can determine traits such as postnatal growth, feed efficiency, milk yield, carcass composition, animal welfare and reproductive potential. We consider the role of epigenetic and cytoplasmic mechanisms of inheritance, and discuss implications for livestock production and future research endeavours. We conclude that although the concept is proven for several traits, issues relating to effect size, and hence commercial importance, remain. Studies have also invariably been conducted under controlled experimental conditions, frequently assessing single risk factors, thereby limiting their translational value for livestock production. We propose concerted international research efforts that consider multiple, concurrent stressors to better represent effects of contemporary animal production systems.
Collapse
|
20
|
Ikeda N, Nakayama Y, Nakazawa N, Yoshida A, Ninomiya H, Shirayoshi Y. Prion Protein and Stage Specific Embryo Antigen 1 as Selection Markers to Enrich the Fraction of Murine Embryonic Stem Cell-Derived Cardiomyocytes. Yonago Acta Med 2016; 59:126-134. [PMID: 27493483 PMCID: PMC4973018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 03/15/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND The prion protein (PrP) might be useful as a tool to collect cardiac progenitor cells derived from embryonic stem (ES) cells. It is also possible that PrP(+) cells include undifferentiated cells with a capacity to develop into tumors. METHODS PrP(+) cells isolated from embryoid bodies (EB) formed by mouse AB1 ES cells were examined using RT-PCR analysis and clonogeneic cell assay. To assess their potential to differentiate into cardiomyocytes, Nkx2.5(GFP/+) (hcgp7) cells, another ES cell line that carries the GFP reporter gene in the Nkx2.5 loci, were used. RESULTS PrP(+) cells isolated from EB of day 7 and 14 did not express pluripotency markers, but expressed cardiac cell markers, while PrP(+) cells isolated from EB of day 21 expressed pluripotency markers. Cultured PrP(+) cells isolated from EB of day 21 expressed pluripotency markers to form colonies, whereas those isolated from EB of day 7 and 14 did not. To exclude proliferating cells from PrP(+) cells, stage specific embryo antigen 1 (SSEA1) was employed as a second marker. PrP(+)/SSEA1(-) cells did not proliferate and expressed cardiac cell markers, while PrP(+)/SSEA1(+) did proliferate. CONCLUSION PrP(+) cells isolated from EB included undifferentiated cells in day 21. PrP(+)/SSEA1(-) cells included cardiomyoctes, suggesting PrP and SSEA1 may be useful as markers to enrich the fraction of cardiomyocytes.
Collapse
Affiliation(s)
- Nobuhito Ikeda
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Tottori University Graduate School of Medical Science, Yonago, 683-8503, Japan
| | - Yuji Nakayama
- †Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University, Yonago, 683-8503, Japan
| | - Natsumi Nakazawa
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Tottori University Graduate School of Medical Science, Yonago, 683-8503, Japan
| | - Akio Yoshida
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Tottori University Graduate School of Medical Science, Yonago, 683-8503, Japan
| | - Haruaki Ninomiya
- ‡Department of Biological Regulation, School of Health Sciences, Tottori University Faculty of Medicine, Yonago, 683-8503, Japan
| | - Yasuaki Shirayoshi
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Tottori University Graduate School of Medical Science, Yonago, 683-8503, Japan
| |
Collapse
|
21
|
Cellular Prion Protein Promotes Neuronal Differentiation of Adipose-Derived Stem Cells by Upregulating miRNA-124. J Mol Neurosci 2016; 59:48-55. [PMID: 26947028 DOI: 10.1007/s12031-016-0733-8] [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: 01/06/2016] [Accepted: 02/23/2016] [Indexed: 10/22/2022]
Abstract
The cellular prion protein (PrP(C)) is a highly conserved glycoprotein anchored by glycosylphosphatidylinositol (GPI) to the cell surface and is also the source of pathogenic agent of scrapie prion protein (PrP(Sc)). Numerous researches have suggested putative physiological roles for PrP(C), including protection from ischemic and excitotoxic lesions, and participation in cell signaling and differentiation. Here, we demonstrated that PrP(C) positively regulates neuronal differentiation of mouse adipose-derived stem cells (ADSCs). The small C-terminal domain phosphatase 1 (SCP1) expression was knocked down by gene silencing. The mRNA expression of miRNA-124 and PrP(C) was measured with quantitative PCR. Western blot analysis was used to detect the protein levels of nestin, βIII-tubulin, and SCP1, and dual-luciferase reporter assay was performed to test the target of miRNA-124. The expression level of PrP(C) was found to increase steadily during neuron-like differentiation process, and PrP(C) knockout resulted in the reduction of neuron-like cell markers. We further showed that miRNA-124 could directly target SCP1-3'-untranslated region to decrease small C-terminal domain phosphatase 1 (SCP1) SCP1, and that miRNA-124 expression is regulated by PrP(C). Our results suggest that PrP(C) may play a key role in the neuronal differentiation of ADSC through modulating miRNA-124-SCP1 axis. To date, this is the first time strong evidence for the involvement of PrP(C) in the neuronal differentiation of ADSC is reported.
Collapse
|
22
|
Halliez S, Martin-Lannerée S, Passet B, Hernandez-Rapp J, Castille J, Urien C, Chat S, Laude H, Vilotte JL, Mouillet-Richard S, Béringue V. Prion protein localizes at the ciliary base during neural and cardiovascular development, and its depletion affects α-tubulin post-translational modifications. Sci Rep 2015; 5:17146. [PMID: 26679898 PMCID: PMC4683536 DOI: 10.1038/srep17146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 10/26/2015] [Indexed: 12/23/2022] Open
Abstract
Although conversion of the cellular form of the prion protein (PrPC) into a misfolded isoform is the underlying cause of prion diseases, understanding PrPC physiological functions has remained challenging. PrPC depletion or overexpression alters the proliferation and differentiation properties of various types of stem and progenitor cells in vitro by unknown mechanisms. Such involvement remains uncertain in vivo in the absence of any drastic phenotype of mice lacking PrPC. Here, we report PrPC enrichment at the base of the primary cilium in stem and progenitor cells from the central nervous system and cardiovascular system of developing mouse embryos. PrPC depletion in a neuroepithelial cell line dramatically altered key cilium-dependent processes, such as Sonic hedgehog signalling and α-tubulin post-translational modifications. These processes were also affected over a limited time window in PrPC–ablated embryos. Thus, our study reveals PrPC as a potential actor in the developmental regulation of microtubule dynamics and ciliary functions.
Collapse
Affiliation(s)
- Sophie Halliez
- INRA (Institut National de la Recherche Agronomique), UR892, Virologie Immunologie Moléculaires, Jouy-en-Josas, France
| | | | - Bruno Passet
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, Jouy-en-Josas, France
| | | | - Johan Castille
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, Jouy-en-Josas, France
| | - Céline Urien
- INRA (Institut National de la Recherche Agronomique), UR892, Virologie Immunologie Moléculaires, Jouy-en-Josas, France
| | - Sophie Chat
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, Jouy-en-Josas, France.,INRA, Plateforme MIMA2, Jouy-en-Josas, France
| | - Hubert Laude
- INRA (Institut National de la Recherche Agronomique), UR892, Virologie Immunologie Moléculaires, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, Jouy-en-Josas, France
| | | | - Vincent Béringue
- INRA (Institut National de la Recherche Agronomique), UR892, Virologie Immunologie Moléculaires, Jouy-en-Josas, France
| |
Collapse
|
23
|
Cellular prion protein promotes post-ischemic neuronal survival, angioneurogenesis and enhances neural progenitor cell homing via proteasome inhibition. Cell Death Dis 2015; 6:e2024. [PMID: 26673668 PMCID: PMC4720898 DOI: 10.1038/cddis.2015.365] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 10/30/2015] [Accepted: 11/16/2015] [Indexed: 12/27/2022]
Abstract
Although cellular prion protein (PrPc) has been suggested to have physiological roles in neurogenesis and angiogenesis, the pathophysiological relevance of both processes remain unknown. To elucidate the role of PrPc in post-ischemic brain remodeling, we herein exposed PrPc wild type (WT), PrPc knockout (PrP−/−) and PrPc overexpressing (PrP+/+) mice to focal cerebral ischemia followed by up to 28 days reperfusion. Improved neurological recovery and sustained neuroprotection lasting over the observation period of 4 weeks were observed in ischemic PrP+/+ mice compared with WT mice. This observation was associated with increased neurogenesis and angiogenesis, whereas increased neurological deficits and brain injury were noted in ischemic PrP−/− mice. Proteasome activity and oxidative stress were increased in ischemic brain tissue of PrP−/− mice. Pharmacological proteasome inhibition reversed the exacerbation of brain injury induced by PrP−/−, indicating that proteasome inhibition mediates the neuroprotective effects of PrPc. Notably, reduced proteasome activity and oxidative stress in ischemic brain tissue of PrP+/+ mice were associated with an increased abundance of hypoxia-inducible factor 1α and PACAP-38, which are known stimulants of neural progenitor cell (NPC) migration and trafficking. To elucidate effects of PrPc on intracerebral NPC homing, we intravenously infused GFP+ NPCs in ischemic WT, PrP−/− and PrP+/+ mice, showing that brain accumulation of GFP+ NPCs was greatly reduced in PrP−/− mice, but increased in PrP+/+ animals. Our results suggest that PrPc induces post-ischemic long-term neuroprotection, neurogenesis and angiogenesis in the ischemic brain by inhibiting proteasome activity.
Collapse
|
24
|
Collins SJ, Tumpach C, Li QX, Lewis V, Ryan TM, Roberts B, Drew SC, Lawson VA, Haigh CL. The prion protein regulates beta-amyloid-mediated self-renewal of neural stem cells in vitro. Stem Cell Res Ther 2015; 6:60. [PMID: 25884827 PMCID: PMC4435829 DOI: 10.1186/s13287-015-0067-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/27/2015] [Accepted: 03/25/2015] [Indexed: 11/10/2022] Open
Abstract
The beta-amyloid (Aβ) peptide and the Aβ-oligomer receptor, prion protein (PrP), both influence neurogenesis. Using in vitro murine neural stem cells (NSCs), we investigated whether Aβ and PrP interact to modify neurogenesis. Aβ imparted PrP-dependent changes on NSC self-renewal, with PrP-ablated and wild-type NSCs displaying increased and decreased cell growth, respectively. In contrast, differentiation of Aβ-treated NSCs into mature cells was unaffected by PrP expression. Such marked PrP-dependent differences in NSC growth responses to Aβ provides further evidence of biologically significant interactions between these two factors and an important new insight into regulation of NSC self-renewal in vivo.
Collapse
Affiliation(s)
- Steven J Collins
- Department of Pathology, The University of Melbourne, Melbourne Brain Centre, Melbourne, VIC, 3010, Australia. .,Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3010, Australia. .,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Carolin Tumpach
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Qiao-Xin Li
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Victoria Lewis
- Department of Pathology, The University of Melbourne, Melbourne Brain Centre, Melbourne, VIC, 3010, Australia.
| | - Timothy M Ryan
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Blaine Roberts
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Simon C Drew
- The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Victoria A Lawson
- Department of Pathology, The University of Melbourne, Melbourne Brain Centre, Melbourne, VIC, 3010, Australia.
| | - Cathryn L Haigh
- Department of Pathology, The University of Melbourne, Melbourne Brain Centre, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
25
|
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
|
26
|
Omatsu-Kanbe M, Nishino Y, Nozuchi N, Sugihara H, Matsuura H. Prion protein- and cardiac troponin T-marked interstitial cells from the adult myocardium spontaneously develop into beating cardiomyocytes. Sci Rep 2014; 4:7301. [PMID: 25466571 PMCID: PMC4252891 DOI: 10.1038/srep07301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/17/2014] [Indexed: 12/22/2022] Open
Abstract
Atypically-shaped cardiomyocytes (ACMs) constitute a novel subpopulation of beating heart cells found in the cultures of cardiac myocyte-removed crude fraction cells obtained from adult mouse cardiac ventricles. Although ~500 beating ACMs are observed under microscope in the cell cultures obtained from the hearts of either male or female mice, the origin of these cells in cardiac tissue has yet to be elucidated due to the lack of exclusive markers. In the present study, we demonstrate the efficacy of cellular prion protein (PrP) as a surface marker of ACMs. Cells expressing PrP at the plasma membrane in the culture of the crude fraction cells were found to develop into beating ACMs by themselves or fuse with each other to become larger multinuclear beating ACMs. Combining PrP with a cardiac-specific contractile protein cardiac troponin T (cTnT) allowed us to identify native ACMs in the mouse cardiac ventricles as either clustered or solitary cells. PrP- and cTnT-marked cells were also found in the adult, even aged, human cardiac ventricles. These findings suggest that interstitial cells marked by PrP and cTnT, native ACMs, exhibit life-long survival in the cardiac ventricles of both mice and humans.
Collapse
Affiliation(s)
- Mariko Omatsu-Kanbe
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192 Japan
| | - Yuka Nishino
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192 Japan
| | - Nozomi Nozuchi
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192 Japan
| | - Hiroyuki Sugihara
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, 520-2192 Japan
| | - Hiroshi Matsuura
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192 Japan
| |
Collapse
|
27
|
Lee YJ, Baskakov IV. The cellular form of the prion protein guides the differentiation of human embryonic stem cells into neuron-, oligodendrocyte-, and astrocyte-committed lineages. Prion 2014; 8:266-75. [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
|
28
|
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
|
29
|
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
|
30
|
Dueñas F, Becerra V, Cortes Y, Vidal S, Sáenz L, Palomino J, De Los Reyes M, Peralta OA. Hepatogenic and neurogenic differentiation of bone marrow mesenchymal stem cells from abattoir-derived bovine fetuses. BMC Vet Res 2014; 10:154. [PMID: 25011474 PMCID: PMC4098697 DOI: 10.1186/1746-6148-10-154] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 06/19/2014] [Indexed: 01/01/2023] Open
Abstract
Background Mesenchymal stem cells (MSC) are multipotent progenitor cells characterized by their ability to both self-renew and differentiate into tissues of mesodermal origin. The plasticity or transdifferentiation potential of MSC is not limited to mesodermal derivatives, since under appropriate cell culture conditions and stimulation by bioactive factors, MSC have also been differentiated into endodermal (hepatocytes) and neuroectodermal (neurons) cells. The potential of MSC for hepatogenic and neurogenic differentiation has been well documented in different animal models; however, few reports are currently available on large animal models. In the present study we sought to characterize the hepatogenic and neurogenic differentiation and multipotent potential of bovine MSC (bMSC) isolated from bone marrow (BM) of abattoir-derived fetuses. Results Plastic-adherent bMSC isolated from fetal BM maintained a fibroblast-like morphology under monolayer culture conditions. Flow cytometric analysis demonstrated that bMSC populations were positive for MSC markers CD29 and CD73 and pluripotency markers OCT4 and NANOG; whereas, were negative for hematopoietic markers CD34 and CD45. Levels of mRNA of hepatic genes α-fetoprotein (AFP), albumin (ALB), alpha1 antitrypsin (α1AT), connexin 32 (CNX32), tyrosine aminotransferase (TAT) and cytochrome P450 (CYP3A4) were up-regulated in bMSC during a 28-Day period of hepatogenic differentiation. Functional analyses in differentiated bMSC cultures evidenced an increase (P < 0.05) in albumin and urea production and glycogen storage. bMSC cultured under neurogenic conditions expressed NESTIN and MAP2 proteins at 24 h of culture; whereas, at 144 h also expressed TRKA and PrPC. Levels of MAP2 and TRKA mRNA were up-regulated at the end of the differentiation period. Conversely, bMSC expressed lower levels of NANOG mRNA during both hepatogenic and neurogenic differentiation processes. Conclusion The expression patterns of linage-specific markers and the production of functional metabolites support the potential for hepatogenic and neurogenic differentiation of bMSC isolated from BM of abattoir-derived fetuses. The simplicity of isolation and the potential to differentiate into a wide variety of cell lineages lays the foundation for bMSC as an interesting alternative for investigation in MSC biology and eventual applications for regenerative therapy in veterinary medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Oscar A Peralta
- Departamento de Fomento de la Producción Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
31
|
Johnson ML, Grazul-Bilska AT, Reynolds LP, Redmer DA. Prion (PrPC) expression in ovine uteroplacental tissues increases after estrogen treatment of ovariectomized ewes and during early pregnancy. Reproduction 2014; 148:1-10. [PMID: 24664411 DOI: 10.1530/rep-13-0548] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Scrapie in sheep is spread laterally by placental transmission of an infectious misfolded form (PrPSc) of a normal prion protein (PrPC) used as a template in PrPSc formation. We hypothesized that PrPC would be expressed in uterine and placental tissues and estradiol-17β (E2) would affect uterine PrPC expression. PrPC expression was evaluated in the uterus of long-term ovariectomized (OVX) ewes treated with an E2 implant for 2-24 h and in uteroplacental tissues from day 20 to day 30 of pregnancy. Expression of PrPC mRNA and PrPC protein increased in the uterus after E2 treatment of OVX ewes. In the maternal placenta, expression of PrPC mRNA and PrPC protein were unchanged, but in the fetal membranes (FM) PrPC mRNA and PrPC protein expression increased from day 20 to day 28. In the nonpregnant uterus, PrPC protein was immunolocalized at apical borders of the surface epithelium, in outer smooth muscle layers of large blood vessels, and in scattered stromal cells of the deep intercaruncular areas of the uterus. In the maternal placenta, PrPC protein was immunolocalized in the cytoplasm of flattened luminal epithelial cells apposed to the FM, whereas in the FM PrPC protein was in trophoblast cells and was also in several tissues of the developing embryo during early pregnancy. These data linking estrogen stimulation to increases in PrPC expression in uteroplacental tissues suggest that PrPC has a specific function during the estrous cycle and early pregnancy. Future studies should determine whether or not estrogen influences PrPC expression in other tissues, such as the nervous system and brain.
Collapse
Affiliation(s)
- Mary Lynn Johnson
- Department of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USADepartment of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USA
| | - Anna T Grazul-Bilska
- Department of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USADepartment of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USA
| | - Lawrence P Reynolds
- Department of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USADepartment of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USA
| | - Dale A Redmer
- Department of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USADepartment of Animal SciencesCenter for Nutrition and PregnancyNorth Dakota State University, Fargo, North Dakota 58108, USA
| |
Collapse
|
32
|
Miranda A, Ramos-Ibeas P, Pericuesta E, Ramirez MA, Gutierrez-Adan A. The role of prion protein in stem cell regulation. Reproduction 2013; 146:R91-9. [PMID: 23740082 DOI: 10.1530/rep-13-0100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cellular prion protein (PrP(C)) has been well described as an essential partner of prion diseases due to the existence of a pathological conformation (PrP(Sc)). Recently, it has also been demonstrated that PrP(C) is an important element of the pluripotency and self-renewal matrix, with an increasing amount of evidence pointing in this direction. Here, we review the data that demonstrate its role in the transcriptional regulation of pluripotency, in the differentiation of stem cells into different lineages (e.g. muscle and neurons), in embryonic development, and its involvement in reproductive cells. Also highlighted are recent results from our laboratory that describe an important regulation by PrP(C) of the major pluripotency gene Nanog. Together, these data support the appearance of new strategies to control stemness, which could represent an important advance in the field of regenerative medicine.
Collapse
Affiliation(s)
- A Miranda
- Departamento de Reproducción Animal, INIA, Avenida Puerta de Hierro no. 12, Local 10, Madrid 28040, Spain
| | | | | | | | | |
Collapse
|
33
|
Peralta OA, Huckle WR, Eyestone WH. Developmental expression of the cellular prion protein (PrP(C) ) in bovine embryos. Mol Reprod Dev 2013; 79:488-98. [PMID: 22674901 DOI: 10.1002/mrd.22057] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The mammalian cellular prion protein (PrP(C) ) is a highly conserved glycoprotein that may undergo conversion into a conformationally altered isoform (scrapie prion protein or PrP(Sc) ), widely believed to be the pathogenic agent of transmissible spongiform encephalopathies (TSEs). Although much is known about PrP(Sc) conversion and its role in TSEs, the normal function of PrP(C) has not been elucidated. In adult mammals, PrP(C) is most abundant in the central nervous tissue, with intermediate levels in the intestine and heart, and lower levels in the pancreas and liver. PrP(C) is expressed during neurogenesis throughout development, and it has recently been proposed that PrP(C) participates in neural cell differentiation during embryogenesis. In order to establish the developmental timing and to address the cell-specific expression of PrP(C) during mammalian development, we examined PrP(C) expression in bovine gametes and embryos through gestation Day 39. Our data revealed differential levels of Prnp mRNA at Days 4 and 18 in pre-attachment embryos. PrP(C) was detected in the developing central and peripheral nervous systems in Day-27, 32-, and -39 embryos. PrP(C) was particularly expressed in differentiated neural cells located in the marginal regions of the central nervous system, but was absent from mitotically active, periventricular areas. Moreover, a PrP(C) cell-specific pattern of expression was detected in non-nervous tissues, including liver and mesonephros, during these stages. The potential participation of PrP(C) in neural cell differentiation is supported by its specific expression in differentiated states of neurogenesis.
Collapse
Affiliation(s)
- Oscar A Peralta
- Department of Large Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| | | | | |
Collapse
|
34
|
Lee YJ, Baskakov IV. The cellular form of the prion protein is involved in controlling cell cycle dynamics, self-renewal, and the fate of human embryonic stem cell differentiation. J Neurochem 2012; 124:310-22. [PMID: 22860629 DOI: 10.1111/j.1471-4159.2012.07913.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 08/02/2012] [Accepted: 08/03/2012] [Indexed: 11/27/2022]
Abstract
Prion protein (PrP(C) ), is a glycoprotein that is expressed on the cell surface. The current study examines the role of PrP(C) in early human embryogenesis using human embryonic stem cells (hESCs) and tetracycline-regulated lentiviral vectors that up-regulate or suppresses PrP(C) expression. Here, we show that expression of PrP(C) in pluripotent hESCs cultured under self-renewal conditions induced cell differentiation toward lineages of three germ layers. Silencing of PrP(C) in hESCs undergoing spontaneous differentiation altered the dynamics of the cell cycle and changed the balance between the lineages of the three germ layers, where differentiation toward ectodermal lineages was suppressed. Moreover, over-expression of PrP(C) in hESCs undergoing spontaneous differentiation inhibited differentiation toward lineages of all three germ layers and helped to preserve high proliferation activity. These results illustrate that PrP(C) is involved in key activities that dictate the status of hESCs including regulation of cell cycle dynamics, controlling the switch between self-renewal and differentiation, and determining the fate of hESCs differentiation. This study suggests that PrP(C) is at the crossroads of several signaling pathways that regulate the switch between preservation of or departure from the self-renewal state, control cell proliferation activity, and define stem cell fate.
Collapse
Affiliation(s)
- Young Jin Lee
- Center for Biomedical Engineering and Technology and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
35
|
Passet B, Young R, Makhzami S, Vilotte M, Jaffrezic F, Halliez S, Bouet S, Marthey S, Khalifé M, Kanellopoulos-Langevin C, Béringue V, Le Provost F, Laude H, Vilotte JL. Prion protein and Shadoo are involved in overlapping embryonic pathways and trophoblastic development. PLoS One 2012; 7:e41959. [PMID: 22860039 PMCID: PMC3408428 DOI: 10.1371/journal.pone.0041959] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 06/27/2012] [Indexed: 12/24/2022] Open
Abstract
The potential requirement of either the Prion or Shadoo protein for early mouse embryogenesis was recently suggested. However, the current data did not allow to precise the developmental process that was affected in the absence of both proteins and that led to the observed early lethal phenotype. In the present study, using various Prnp transgenic mouse lines and lentiviral vectors expressing shRNAs that target the Shadoo-encoding mRNA, we further demonstrate the specific requirement of at least one of these two PrP-related proteins at early developmental stages. Histological analysis reveals developmental defect of the ectoplacental cone and important hemorrhage surrounding the Prnp-knockout-Sprn-knockdown E7.5 embryos. By restricting the RNA interference to the trophoblastic cell lineages, the observed lethal phenotype could be attributed to the sole role of these proteins in this trophectoderm-derived compartment. RNAseq analysis performed on early embryos of various Prnp and Sprn genotypes indicated that the simultaneous down-regulation of these two proteins affects cell-adhesion and inflammatory pathways as well as the expression of ectoplacental-specific genes. Overall, our data provide biological clues in favor of a crucial and complementary embryonic role of the prion protein family in Eutherians and emphasizes the need to further evaluate its implication in normal and pathological human placenta biology.
Collapse
Affiliation(s)
- Bruno Passet
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Rachel Young
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Samira Makhzami
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Marthe Vilotte
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Florence Jaffrezic
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Sophie Halliez
- INRA, UR892, Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | - Stéphan Bouet
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Sylvain Marthey
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Manal Khalifé
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Colette Kanellopoulos-Langevin
- Laboratory of Inflammation, Gestation and Autoimmunity, J. Monod Institute, UMR 7592 (CNRS and University Diderot), Paris, France
| | - Vincent Béringue
- INRA, UR892, Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | - Fabienne Le Provost
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Hubert Laude
- INRA, UR892, Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
- * E-mail:
| |
Collapse
|
36
|
Human prion protein binds Argonaute and promotes accumulation of microRNA effector complexes. Nat Struct Mol Biol 2012; 19:517-24, S1. [PMID: 22484317 DOI: 10.1038/nsmb.2273] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 02/28/2012] [Indexed: 12/13/2022]
Abstract
Despite intense research in the context of neurodegenerative diseases associated with its misfolding, the endogenous human prion protein PrP(C) (or PRNP) has poorly understood physiological functions. Whereas most PrP(C) is exposed to the extracellular environment, conserved domains result in transmembrane forms of PrP(C) that traffic in the endolysosomal system and are linked to inherited and infectious neuropathologies. One transmembrane PrP(C) variant orients the N-terminal 'octarepeat' domain into the cytoplasm. Here we demonstrate that the octarepeat domain of human PrP(C) contains GW/WG motifs that bind Argonaute (AGO) proteins, the essential components of microRNA (miRNA)-induced silencing complexes (miRISCs). Transmembrane PrP(C) preferentially binds AGO, and PrP(C) promotes formation or stability of miRISC effector complexes containing the trinucleotide repeat-containing gene 6 proteins (TNRC6) and miRNA-repressed mRNA. Accordingly, effective repression of several miRNA targets requires PrP(C). We propose that dynamic interactions between PrP(C)-enriched endosomes and subcellular foci of AGO underpin these effects.
Collapse
|
37
|
Abstract
Prion protein (PrP) can be considered a pivotal molecule because it interacts with several partners to perform a diverse range of critical biological functions that might differ in embryonic and adult cells. In recent years, there have been major advances in elucidating the putative role of PrP in the basic biology of stem cells in many different systems. Here, we review the evidence indicating that PrP is a key molecule involved in driving different aspects of the potency of embryonic and tissue-specific stem cells in self-perpetuation and differentiation in many cell types. It has been shown that PrP is involved in stem cell self-renewal, controlling pluripotency gene expression, proliferation, and neural and cardiomyocyte differentiation. PrP also has essential roles in distinct processes that regulate tissue-specific stem cell biology in nervous and hematopoietic systems and during muscle regeneration. Results from our own investigations have shown that PrP is able to modulate self-renewal and proliferation in neural stem cells, processes that are enhanced by PrP interactions with stress inducible protein 1 (STI1). Thus, the available data reveal the influence of PrP in acting upon the maintenance of pluripotent status or the differentiation of stem cells from the early embryogenesis through adulthood.
Collapse
Affiliation(s)
- Marilene H Lopes
- Department of Cell and Developmental Biology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil.
| | | |
Collapse
|
38
|
Garza MC, Fernández-Borges N, Bolea R, Badiola JJ, Castilla J, Monleón E. Detection of PrPres in genetically susceptible fetuses from sheep with natural scrapie. PLoS One 2011; 6:e27525. [PMID: 22194786 PMCID: PMC3237407 DOI: 10.1371/journal.pone.0027525] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 10/18/2011] [Indexed: 11/19/2022] Open
Abstract
Scrapie is a transmissible spongiform encephalopathy with a wide PrPres dissemination in many non-neural tissues and with high levels of transmissibility within susceptible populations. Mechanisms of transmission are incompletely understood. It is generally assumed that it is horizontally transmitted by direct contact between animals or indirectly through the environment, where scrapie can remain infectious for years. In contrast, in utero vertical transmission has never been demonstrated and has rarely been studied. Recently, the use of the protein misfolding cyclic amplification technique (PMCA) has allowed prion detection in various tissues and excretions in which PrPres levels have been undetectable by traditional assays. The main goal of this study was to detect PrPres in fetal tissues and the amniotic fluid from natural scrapie infected ewes using the PMCA technique. Six fetuses from three infected pregnant ewes in an advanced clinical stage of the disease were included in the study. From each fetus, amniotic fluid, brain, spleen, ileo-cecal valve and retropharyngeal lymph node samples were collected and analyzed using Western blotting and PMCA. Although all samples were negative using Western blotting, PrPres was detected after in vitro amplification. Our results represent the first time the biochemical detection of prions in fetal tissues, suggesting that the in utero transmission of scrapie in natural infected sheep might be possible.
Collapse
Affiliation(s)
- María Carmen Garza
- Centro de Investigación en Encefalopatías Espongiformes Transmisibles y Enfermedades Emergentes, Universidad de Zaragoza, Zaragoza, Spain
| | | | - Rosa Bolea
- Centro de Investigación en Encefalopatías Espongiformes Transmisibles y Enfermedades Emergentes, Universidad de Zaragoza, Zaragoza, Spain
| | - Juan José Badiola
- Centro de Investigación en Encefalopatías Espongiformes Transmisibles y Enfermedades Emergentes, Universidad de Zaragoza, Zaragoza, Spain
| | - Joaquín Castilla
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Eva Monleón
- Centro de Investigación en Encefalopatías Espongiformes Transmisibles y Enfermedades Emergentes, Universidad de Zaragoza, Zaragoza, Spain
- Producció Animal, Universitat de Lleida, LLeida, Spain
- * E-mail:
| |
Collapse
|
39
|
Santos TG, Silva IR, Costa-Silva B, Lepique AP, Martins VR, Lopes MH. Enhanced neural progenitor/stem cells self-renewal via the interaction of stress-inducible protein 1 with the prion protein. Stem Cells 2011; 29:1126-36. [PMID: 21608082 DOI: 10.1002/stem.664] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Prion protein (PrP(C) ), when associated with the secreted form of the stress-inducible protein 1 (STI1), plays an important role in neural survival, neuritogenesis, and memory formation. However, the role of the PrP(C) -STI1 complex in the physiology of neural progenitor/stem cells is unknown. In this article, we observed that neurospheres cultured from fetal forebrain of wild-type (Prnp(+/+) ) and PrP(C) -null (Prnp(0/0) ) mice were maintained for several passages without the loss of self-renewal or multipotentiality, as assessed by their continued capacity to generate neurons, astrocytes, and oligodendrocytes. The homogeneous expression and colocalization of STI1 and PrP(C) suggest that they may associate and function as a complex in neurosphere-derived stem cells. The formation of neurospheres from Prnp(0/0) mice was reduced significantly when compared with their wild-type counterparts. In addition, blockade of secreted STI1, and its cell surface ligand, PrP(C) , with specific antibodies, impaired Prnp(+/+) neurosphere formation without further impairing the formation of Prnp(0/0) neurospheres. Alternatively, neurosphere formation was enhanced by recombinant STI1 application in cells expressing PrP(C) but not in cells from Prnp(0/0) mice. The STI1-PrP(C) interaction was able to stimulate cell proliferation in the neurosphere-forming assay, while no effect on cell survival or the expression of neural markers was observed. These data suggest that the STI1-PrP(C) complex may play a critical role in neural progenitor/stem cells self-renewal via the modulation of cell proliferation, leading to the control of the stemness capacity of these cells during nervous system development.
Collapse
Affiliation(s)
- Tiago G Santos
- Department of Molecular and Cell Biology, International Center for Research and Education, Antonio Prudente Foundation, A. C. Camargo Hospital and National Institute for Translational Neuroscience (CNPq/MCT), São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
40
|
Khalifé M, Young R, Passet B, Halliez S, Vilotte M, Jaffrezic F, Marthey S, Béringue V, Vaiman D, Le Provost F, Laude H, Vilotte JL. Transcriptomic analysis brings new insight into the biological role of the prion protein during mouse embryogenesis. PLoS One 2011; 6:e23253. [PMID: 21858045 PMCID: PMC3156130 DOI: 10.1371/journal.pone.0023253] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 07/11/2011] [Indexed: 11/25/2022] Open
Abstract
The biological function of the Prion protein remains largely unknown but recent data revealed its implication in early zebrafish and mammalian embryogenesis. To gain further insight into its biological function, comparative transcriptomic analysis between FVB/N and FVB/N Prnp knockout mice was performed at early embryonic stages. RNAseq analysis revealed the differential expression of 73 and 263 genes at E6.5 and E7.5, respectively. The related metabolic pathways identified in this analysis partially overlap with those described in PrP1 and PrP2 knockdown zebrafish embryos and prion-infected mammalian brains and emphasize a potentially important role for the PrP family genes in early developmental processes.
Collapse
Affiliation(s)
- Manal Khalifé
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Rachel Young
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Bruno Passet
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Sophie Halliez
- INRA, UR892, Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | - Marthe Vilotte
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Florence Jaffrezic
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Sylvain Marthey
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Vincent Béringue
- INRA, UR892, Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | | | - Fabienne Le Provost
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
| | - Hubert Laude
- INRA, UR892, Virologie et Immunologie Moléculaires, INRA, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, INRA, Jouy-en-Josas, France
- * E-mail:
| |
Collapse
|