1
|
Dalai W, Matsuo E, Takeyama N, Kawano J, Saeki K. CpG site DNA methylation patterns reveal a novel regulatory element in the mouse prion protein gene. J Vet Med Sci 2017; 79:100-107. [PMID: 27666463 PMCID: PMC5289245 DOI: 10.1292/jvms.16-0390] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The cellular isoform of the prion protein (PrPC) plays critical roles in the development of prion disorders. Although PrP mRNA is ubiquitously
present in a tissue-specific manner, the DNA methylation of PrP gene (Prnp) is still unknown. In this study, we demonstrated that the CpG
island (CGI, positioned at −218 to +152 bp from the transcriptional start site) including the Prnp core promoter region was completely
unmethylated in all tested tissues. On the other hand, CpG methylation in the CGI shore region (positioned at −599 to −238 bp) occurred in various tissue- and
site-specific proportions. Interestingly, the correlation analysis between CpG methylation status and PrP mRNA levels showed that one CpG site methylation at
−576 was negatively correlated with the PrP mRNA level (Pearson’s r = −0.374, P=0.035). Taken together, our results suggest
that Prnp is a typical housekeeping gene and various methylation frequencies of the CGI shore region are likely to affect Prnp
expression in a tissue-specific manner.
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
|
2
|
Deletion of the prion gene Prnp affects offensive aggression in mice. Behav Brain Res 2014; 266:216-21. [DOI: 10.1016/j.bbr.2014.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 02/27/2014] [Accepted: 03/03/2014] [Indexed: 01/06/2023]
|
3
|
Miki T, Yokoyama T, Kusaka T, Suzuki S, Ohta KI, Warita K, Wang ZY, Ueki M, Sumitani K, Bellinger FP, Tamai M, Liu JQ, Yakura T, Takeuchi Y. Early postnatal repeated maternal deprivation causes a transient increase in OMpg and BDNF in rat cerebellum suggesting precocious myelination. J Neurol Sci 2014; 336:62-7. [DOI: 10.1016/j.jns.2013.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 09/20/2013] [Accepted: 10/04/2013] [Indexed: 10/26/2022]
|
4
|
Lim ST, Chang A, Giuliano RE, Federoff HJ. Ectodomain shedding of nectin-1 regulates the maintenance of dendritic spine density. J Neurochem 2011; 120:741-51. [PMID: 22118475 DOI: 10.1111/j.1471-4159.2011.07592.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Synaptic remodeling has been postulated as a mechanism underlying synaptic plasticity and cell adhesion molecules are thought to contribute to this process. We examined the role of nectin-1 ectodomain shedding on synaptogenesis in cultured rat hippocampal neurons. Nectins are Ca(2+) -independent immunoglobulin-like adhesion molecules, involved in cell-cell adherens junctions. Herein, we show that the processing of nectin-1 occurs by multiple endoproteolytic steps both in vivo and in vitro. We identified regions containing two distinct cleavage sites within the ectodomain of nectin-1. By alanine scanning mutagenesis, two point mutations that disrupt nectin-1 ectodomain cleavage events were identified. Expression of these mutants significantly alters the density of dendritic spines. These findings suggest that ectodomain shedding of nectin-1 regulates dendritic spine density and related synaptic functions.
Collapse
Affiliation(s)
- Seung T Lim
- Neuroscience Department, Georgetown University Medical Center, Washington, District of Columbia 20057, USA
| | | | | | | |
Collapse
|
5
|
Benvegnù S, Roncaglia P, Agostini F, Casalone C, Corona C, Gustincich S, Legname G. Developmental influence of the cellular prion protein on the gene expression profile in mouse hippocampus. Physiol Genomics 2011; 43:711-25. [PMID: 21406608 DOI: 10.1152/physiolgenomics.00205.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The conversion of the cellular prion protein (PrP(C)) to an abnormal and protease-resistant isoform is the key event in prion diseases. Mice lacking PrP(C) are resistant to prion infection, and downregulation of PrP(C) during prion infection prevents neuronal loss and the progression to clinical disease. These results are suggestive of the potential beneficial effect of silencing PrP(C) during prion diseases. However, the silencing of a protein that is widely expressed throughout the central nervous system could be detrimental to brain homeostasis. The physiological role of PrP(C) remains still unclear, but several putative functions (e.g., neuronal development and maintenance) have been proposed. To assess the influence of PrP(C) on gene expression profile in the mouse brain, we undertook a microarray analysis by using RNA isolated from the hippocampus at two different developmental stages: newborn (4.5-day-old) and adult (3-mo-old) mice, both from wild-type and Prnp(0/0) animals. Comparing the different datasets allowed us to identify "commonly" co-regulated genes and "uniquely" deregulated genes during postnatal development. The absence of PrP(C) affected several biological pathways, the most representative being cell signaling, cell-cell communication and transduction processes, calcium homeostasis, nervous system development, synaptic transmission, and cell adhesion. However, there was only a moderate alteration of the gene expression profile in our animal models. PrP(C) deficiency did not lead to a dramatic alteration of gene expression profile and produced moderately altered gene expression levels from young to adult animals. Thus, our results may provide additional support to silencing endogenous PrP(C) levels as therapeutic approach to prion diseases.
Collapse
Affiliation(s)
- Stefano Benvegnù
- Laboratory of Prion Biology, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste
| | | | | | | | | | | | | |
Collapse
|
6
|
Irmady K, Zechel S, Unsicker K. Fibroblast growth factor 2 regulates astrocyte differentiation in a region-specific manner in the hindbrain. Glia 2011; 59:708-19. [DOI: 10.1002/glia.21141] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 12/15/2010] [Indexed: 01/04/2023]
|
7
|
Benvegnù S, Poggiolini I, Legname G. Neurodevelopmental expression and localization of the cellular prion protein in the central nervous system of the mouse. J Comp Neurol 2010; 518:1879-91. [PMID: 20394048 DOI: 10.1002/cne.22357] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs) are neurodegenerative disorders caused by PrP(Sc), or prion, an abnormally folded form of the cellular prion protein (PrP(C)). The abundant expression of PrP(C) in the central nervous system (CNS) is a requirement for prion replication, yet despite years of intensive research the physiological function of PrP(C) still remains unclear. Several routes of investigation point out a potential role for PrP(C) in axon growth and neuronal development. Thus, we undertook a detailed analysis of the spatial and temporal expression of PrP(C) during mouse CNS development. Our findings show regional differences of the expression of PrP, with some specific white matter structures showing the earliest and highest expression of PrP(C). Indeed, all these regions are part of the thalamolimbic neurocircuitry, suggesting a potential role of PrP(C) in the development and functioning of this specific brain system.
Collapse
Affiliation(s)
- Stefano Benvegnù
- Scuola Internazionale Superiore di Studi Avanzati-International School for Advanced Studies (SISSA-ISAS), Neurobiology Sector, I-34151 Trieste, Italy
| | | | | |
Collapse
|
8
|
Linden R, Martins VR, Prado MAM, Cammarota M, Izquierdo I, Brentani RR. Physiology of the prion protein. Physiol Rev 2008; 88:673-728. [PMID: 18391177 DOI: 10.1152/physrev.00007.2007] [Citation(s) in RCA: 435] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prion diseases are transmissible spongiform encephalopathies (TSEs), attributed to conformational conversion of the cellular prion protein (PrP(C)) into an abnormal conformer that accumulates in the brain. Understanding the pathogenesis of TSEs requires the identification of functional properties of PrP(C). Here we examine the physiological functions of PrP(C) at the systemic, cellular, and molecular level. Current data show that both the expression and the engagement of PrP(C) with a variety of ligands modulate the following: 1) functions of the nervous and immune systems, including memory and inflammatory reactions; 2) cell proliferation, differentiation, and sensitivity to programmed cell death both in the nervous and immune systems, as well as in various cell lines; 3) the activity of numerous signal transduction pathways, including cAMP/protein kinase A, mitogen-activated protein kinase, phosphatidylinositol 3-kinase/Akt pathways, as well as soluble non-receptor tyrosine kinases; and 4) trafficking of PrP(C) both laterally among distinct plasma membrane domains, and along endocytic pathways, on top of continuous, rapid recycling. A unified view of these functional properties indicates that the prion protein is a dynamic cell surface platform for the assembly of signaling modules, based on which selective interactions with many ligands and transmembrane signaling pathways translate into wide-range consequences upon both physiology and behavior.
Collapse
Affiliation(s)
- Rafael Linden
- Instituto de Biofísica da Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | |
Collapse
|
9
|
Salimi K, Glantz LA, Hamer RM, German TT, Gilmore JH, Jarskog LF. Regulation of complexin 1 and complexin 2 in the developing human prefrontal cortex. Synapse 2008; 62:273-82. [PMID: 18240322 DOI: 10.1002/syn.20492] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Complexin 1 (CX1) and complexin 2 (CX2) are presynaptic proteins that modulate neurotransmitter release and are used as markers of inhibitory and excitatory synapses, respectively. The aim of this study was to gain insight into the development of inhibitory and excitatory synapses in human prefrontal cortex (PFC) by examining the expression of CX1 and CX2 in postmortem tissues. Relative complexin protein levels were measured by Western blotting in postmortem dorsolateral prefrontal cortex (DLPFC) of 42 subjects without neurological or psychiatric disease ranging in age from 18 gestational weeks to 25 years. Samples were batched a priori into fetal, 0-12 month, 1-5 years, 6-10 years, 11-15 years, 16-20 years, and 21-25 years age groups. CX1 and CX2 expression and CX2/CX1 demonstrated a significant effect of age group by ANOVA. Group CX1 level increased progressively across development and was lowest in the fetal group and highest in the young adult group, whereas group CX2 level increased between the fetal and the 6-10 years groups and then plateaued. Consistent with these divergent patterns, there was a significant effect of age group on CX2/CX1, which was higher in fetal and infant groups than in the young adult group. Furthermore, regression analysis demonstrated linear relationships of CX1 and CX2/CX1 with age, whereas CX2 was better described as having a curvilinear relationship with age. These data indicate that complexin expression increases during synaptic maturation in human DLPFC and that an increase in the influence of inhibitory synapses relative to that of excitatory synapses occurs during development in this cortical region.
Collapse
Affiliation(s)
- Kayvon Salimi
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | | | | | | |
Collapse
|
10
|
Witusik M, Gresner SM, Hulas-Bigoszewska K, Krynska B, Azizi SA, Liberski PP, Brown P, Rieske P. Neuronal and astrocytic cells, obtained after differentiation of human neural GFAP-positive progenitors, present heterogeneous expression of PrPc. Brain Res 2007; 1186:65-73. [PMID: 17996224 DOI: 10.1016/j.brainres.2007.10.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 09/28/2007] [Accepted: 10/06/2007] [Indexed: 11/19/2022]
Abstract
PrP(c) is a cellular isoform of the prion protein with an unknown normal function. One of the putative physiological roles of this protein is its involvement in cell differentiation. Recently, in vitro and in vivo studies showed that GFAP-positive cells have characteristics of stem/progenitor cells that generate neurons and glia. We used an in vitro model of human neurogenesis from GFAP-positive progenitor cells to study the expression of PrP(c) during neural differentiation. Semi-quantitative multiplex-PCR assay and Western blot analysis revealed a significant increase of PRNP expression level in differentiated cells compared to undifferentiated cell population. As determined by immunocytochemistry followed by a quantitative image analysis, the PrP(c) level increased significantly in neuronal cells and did not increase significantly in glial cells. Of note, glial and neuronal cells showed a very large heterogeneity of PrP(c) expression. Our results provide the basis for studying the role of PrP(c) in cell differentiation and neurogenesis from human GFAP-positive progenitor cells.
Collapse
Affiliation(s)
- Monika Witusik
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Lodz, 8/10 Czechoslowacka str., Lodz, Poland
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Diez M, Groth D, DeArmond SJ, Prusiner SB, Hökfelt T. Changes in neuropeptide expression in mice infected with prions. Neurobiol Aging 2007; 28:748-65. [PMID: 16621165 DOI: 10.1016/j.neurobiolaging.2006.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Revised: 02/24/2006] [Accepted: 02/28/2006] [Indexed: 10/24/2022]
Abstract
Prion diseases are neurodegenerative disorders characterized by accumulation of an aberrantly folded isoform (PrP(Sc)) of the normal prion protein (PrP(C)). Using in situ hybridization and immunohistochemistry, we have studied changes in the expression of neuropeptides, acetylcholinesterase and tyrosine hydroxylase in CD1 and FVB wild-type mouse strains, as well as in PrP(C) null mice and in mice overexpressing PrP(C) following intracerebral inoculation with RML or Me7 prions. In the immunohistochemical analysis, neuropeptide Y (NPY), enkephalin and dynorphin-like immunoreactivities increased in mossy fibers of CD1 and FVB mice inoculated with either RML- or Me7 prions, whereas cholecystokinin-like immunoreactivity was decreased. These changes in peptide levels were paralleled by an increase in the transcripts in granule cells for neuropeptide Y, enkephalin, and cholecystokinin. However, the dynorphin transcript was decreased in the granule cells. The changes occurred more rapidly in PrP(C)-overexpressing compared to wild-type mice, and could not be found at all in PrP(C)-knockout mice. These changes in peptide expression, which mostly occur before appearance of symptoms of disease, may reflect attempts to initiate protective and/or regenerative processes.
Collapse
Affiliation(s)
- Margarita Diez
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
12
|
Adle-Biassette H, Verney C, Peoc'h K, Dauge MC, Razavi F, Choudat L, Gressens P, Budka H, Henin D. Immunohistochemical expression of prion protein (PrPC) in the human forebrain during development. J Neuropathol Exp Neurol 2006; 65:698-706. [PMID: 16825956 DOI: 10.1097/01.jnen.0000228137.10531.72] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The cellular prion protein (PrPC) is a ubiquitous protein whose expression in the adult brain occurs mainly in synapses. We used monoclonal antibodies to study fetal and perinatal PrPC expression in the human forebrain. Double immunofluorescence and confocal microscopy with GFAP, Iba1, MAP2, doublecortin, synaptophysin, and GAP-43 were used to localize PrPC. PrPC immunoreactivity was observed in axonal tracts and fascicles from the 11th week to the end of gestation. Synapses expressed PrPC at increasing levels throughout synaptogenesis. At midgestation, a few PrPC-labeled neurons were detected in the cortical anlage and numerous ameboid and intermediate microglial cells were PrPC-positive. In contrast, at the end of gestation, microglial PrPC expression decreased to almost nothing, whereas neuronal PrPC expression increased, most notably in ischemic areas. In adults, PrPC immunoreactivity was restricted to the synaptic neuropil of the gray matter. At all ages, choroid plexus, ependymal, and endothelial cells were labeled, whereas astrocytes were only occasionally immunoreactive. In conclusion, the early expression of PrPC in the axonal field may suggest a specific role for this molecule in axonal growth during development. Moreover, PrPC may play a role in early microglial cell development.
Collapse
Affiliation(s)
- Homa Adle-Biassette
- AP HP, Hôpital Bichat-Claude Bernard, Service d'Anatomie Pathologie, Université Paris 7, Faculté de Médecine Denis Diderot, Paris, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Oo TF, Ries V, Cho J, Kholodilov N, Burke RE. Anatomical basis of glial cell line-derived neurotrophic factor expression in the striatum and related basal ganglia during postnatal development of the rat. J Comp Neurol 2005; 484:57-67. [PMID: 15717300 PMCID: PMC3092474 DOI: 10.1002/cne.20463] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
There is increasing evidence that glial cell line-derived neurotrophic factor (GDNF) plays a role as a limiting, striatal target-derived neurotrophic factor for dopamine neurons of the substantia nigra pars compacta (SNpc) by regulating the magnitude of the first phase of postnatal natural cell death which occurs in these neurons. While it has been shown that GDNF mRNA is relatively abundant in postnatal striatum, the cellular basis of its expression has been unknown. We therefore used nonradioactive in situ hybridization and immunohistochemistry to examine the cellular basis of GDNF mRNA and protein expression, respectively, in postnatal striatum and related structures. We found that GDNF mRNA is expressed within medium-sized striatal neurons. Expression in glia was not observed. At the protein level, regionally, GDNF expression in striatum was observed in striosomal patches, as previously described. At a cellular level a few neurons were observed, but they do not account for the striosomal pattern. This pattern is predominantly due to GDNF-positive neuropil. Some of this neuropil arises from tyrosine hydroxylase-positive nigro-striatal dopaminergic afferents. Astrocytic processes do not appear to contribute to the striosomal pattern. GDNF-positive fibers are identified not only within intrinsic striatal neuropil, but also in fibers within the major striatal efferent targets: the globus pallidus, the entopeduncular nucleus, and the SN pars reticulata. We conclude that during normal postnatal development, medium-sized neurons are the principal source of GDNF within the striatum.
Collapse
Affiliation(s)
- Tinmarla Frances Oo
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Vincent Ries
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Jinwhan Cho
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Nikolai Kholodilov
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Robert E. Burke
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
- Department of Pathology, The College of Physicians and Surgeons, Columbia University, New York, New York 10032
| |
Collapse
|
14
|
Cho J, Yarygina O, Oo TF, Kholodilov NG, Burke RE. Glial cell line-derived neurotrophic factor receptor GFRα1 is expressed in the rat striatum during postnatal development. ACTA ACUST UNITED AC 2004; 127:96-104. [PMID: 15306125 DOI: 10.1016/j.molbrainres.2004.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2004] [Indexed: 10/26/2022]
Abstract
Dopamine neurons of the substantia nigra (SN) undergo a natural cell death event which is biphasic, with peaks at postnatal days (PNDs) 2 and 14. There is growing evidence that GDNF functions as a striatal target-derived neurotrophic factor to regulate the first phase. It has been unknown whether the GDNF receptor, GFRalpha1, may play a role in regulating either phase. To evaluate a possible role for GFRalpha1 we have examined its expression throughout postnatal development in the SN and particularly in the striatum, where its expression has been uncertain. GFRalpha1 mRNA is highly expressed in SN, as previously shown, with highest levels at PND14-28. We find that it is also expressed in striatum with a similar time course, but with a more discrete period of maximal expression between PND10 and PND14. The cellular basis of this maximum of expression is an increased number of GFRalpha1 mRNA-positive medium-sized neurons evenly distributed within the striatum. Immunostaining reveals GFRalpha1 protein-positive neurons with a similar morphology and distribution. We conclude that GFRalpha1 is expressed in striatum maximally late in postnatal development. In this location it may act in trans to influence the viability and development of nigral dopamine neurons.
Collapse
Affiliation(s)
- JinWhan Cho
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, Room 308, Black Building, 650 West 168th Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
15
|
Knutson M, Menzies S, Connor J, Wessling-Resnick M. Developmental, regional, and cellular expression of SFT/UbcH5A and DMT1 mRNA in brain. J Neurosci Res 2004; 76:633-41. [PMID: 15139022 DOI: 10.1002/jnr.20113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Brain iron has marked developmental, regional, and cellular distribution patterns. To characterize better the potential mechanisms for iron transport into and within the brain, we have analyzed expression patterns of two factors: divalent metal transporter 1 (DMT1) and stimulator of Fe transport (SFT). DMT1 is known to participate in brain iron uptake although functional information is lacking. Even less clear is the possible role of SFT, which is related to a member of the ubiquitin-conjugating E2 family UbcH5A, but previous studies have found SFT/Ubc5Ha mRNA expressed abundantly in mouse brain. Like DMT1, SFT function has been implicated in transferrin and nontransferrin-bound iron uptake. Comparative Northern analysis indicates that SFT/UbcH5A mRNA levels are threefold higher in 3-day-old mice than at later ages, whereas levels of DMT1 mRNA do not change. In situ analysis of neonatal mouse brain reveals prominent SFT/UbcH5A mRNA expression in epithelial and ependymal cells in the choroid plexus and neurons of the olfactory bulb, hippocampus, and cortex. Adult mouse brain expresses SFT/UbcH5A mRNA mainly in white matter of the cerebellum and pons. Using a multiple tissue expression (MTE) array containing 20 different human brain regions, the highest levels of both SFT/UbcH5A and DMT1 mRNA are detected in the corpus callosum and cerebellum. The significantly elevated levels of SFT/UbcH5A mRNA in the neonatal mouse and its localization to choroid plexus, a major site of brain iron acquisition, suggest that this factor may contribute to the rapid rate of brain iron uptake that occurs in the early postnatal period.
Collapse
Affiliation(s)
- Mitchell Knutson
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
16
|
Kühn K, Zhu XR, Lübbert H, Stichel CC. Parkin expression in the developing mouse. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 149:131-42. [PMID: 15063093 DOI: 10.1016/j.devbrainres.2004.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/18/2004] [Indexed: 10/26/2022]
Abstract
Parkin is an E3 ubiquitin ligase causally involved in the pathogenesis of autosomal recessive juvenile parkinsonism. In this paper, we analysed the formation of alternative splice products and the spatio-temporal expression pattern of parkin during pre- and postnatal mouse development. Using RT-PCR, Northern blot, in situ hybridization, Western blot analysis, and immunohistochemistry we found (i) alternative splice forms of parkin; (ii) an early and widespread expression of parkin mRNA and protein in the CNS and several organs, already at E10/12; (iii) a marked increase in expression level during midgestational development (E15-18) in the CNS, followed by a steady increase until adulthood; (iv) an ubiquitous distribution throughout CNS ontogeny. Our results show that parkin expression is correlated with cell maturation and suggests an important physiological role of parkin in neurons that is at no time limited to the dopaminergic system.
Collapse
Affiliation(s)
- Kati Kühn
- Department of Animal Physiology, ND5/132, Ruhr-University of Bochum, D-44780 Bochum, Germany
| | | | | | | |
Collapse
|
17
|
Röhl C, Held-Feindt J, Sievers J. Developmental changes of parameters for astrogliosis during cultivation of purified cerebral astrocytes from newborn rats. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2003; 144:191-9. [PMID: 12935916 DOI: 10.1016/s0165-3806(03)00171-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Astrogliosis is a common phenomenon seen in most neuropathological changes of the central nervous system. Several in vitro models have been used to study the mechanisms and conditions for the induction of astrogliosis, however many do not take into account that the metabolic and structural characteristics of astrocytes change with time in culture. Thus, it appears difficult to attribute changes of, e.g., GFAP to the normal change in vitro as opposed to additional changes due to an astrogliotic reaction. The present study was therefore undertaken to characterize these developmental changes in purified astroglial secondary cultures during cultivation to provide a basis for further investigations of astrogliosis in vitro. During 6 weeks of cultivation (3-43 days) GFAP (ELISA) increased much more (22-fold) than the cell number (2.5-fold) and the total protein (3.5-fold). The GFAP/protein ratio increased during the first 4 weeks of cultivation and reached a plateau thereafter, which was accompanied by a significant increase of GFAP mRNA (Northern blot). At the ultrastructural level (transmission electron microscopy) gliofilaments in the perinuclear region as well as in the cell processes of 4-day-old astrocytes showed a dispersed pattern, whereas an accumulation of gliofilaments was found in 39-day-old cells, which formed large aggregated bundles localized mostly in the cell processes. Our results show that in vitro astrocytes undergo developmental changes in their accumulation of GFAP and intermediate filaments which reach a stable steady state after 4 weeks in culture. These 'normal' developmental changes will have to be taken into account, when experiments with variations of the level of GFAP are performed. Stable culture conditions for experimentation appear to be present after 4 weeks in culture.
Collapse
Affiliation(s)
- Claudia Röhl
- Department of Anatomy, University of Kiel, Olshausenstr 40, D-24098 Kiel, Germany.
| | | | | |
Collapse
|
18
|
Weber P, Schuler M, Gérard C, Mark M, Metzger D, Chambon P. Temporally controlled site-specific mutagenesis in the germ cell lineage of the mouse testis. Biol Reprod 2003; 68:553-9. [PMID: 12533419 DOI: 10.1095/biolreprod.102.005801] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
We have obtained a PrP-Cre-ER(T) transgenic mouse line (28.8) that selectively expresses in testis the tamoxifen-inducible Cre-ER(T) recombinase under the control of a mouse Prion protein (PrP) promoter-containing genomic fragment. Cre-ER(T) is expressed in spermatogonia and spermatocytes, but not in Sertoli and Leydig cells. We also established reporter PrP-L-EGFP-L transgenic mice harboring a LoxP-flanked enhanced green fluorescent protein (EGFP) Cre reporter cassette under the control of the same PrP promoter-containing genomic fragment that exhibits prominent EGFP expression in brain and testis. Using the PrP-L-EGFP-L as well as other Cre-reporter mice, we demonstrate that tamoxifen administration efficiently and selectively induces Cre-mediated recombination in the germ cell lineage. The established PrP-Cre-ER(T) line should provide a valuable tool for studying functions of germ cell-expressed genes involved in spermatogenesis.
Collapse
Affiliation(s)
- Philipp Weber
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université Louis Pasteur, Collège de France, B P 163, 67404 Illkirch-Cedex, Communauté Urbaine de Strasbourg, France
| | | | | | | | | | | |
Collapse
|
19
|
Miele G, Blanco ARA, Baybutt H, Horvat S, Manson J, Clinton M. Embryonic activation and developmental expression of the murine prion protein gene. Gene Expr 2003; 11:1-12. [PMID: 12691521 PMCID: PMC5991155 DOI: 10.3727/000000003783992324] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2002] [Indexed: 02/02/2023]
Abstract
While it is well established that cellular prion protein (PrP(C)) expression is required for the development of transmissible spongiform encephalopathies (TSEs), the physiological function of PrP(C) has yet to be determined. A number of studies have examined PrP expression in different tissues and in the later stages of embryonic development. However, the relative levels of expression of PrP RNA and protein in tissues outside the central nervous system (CNS) is not well documented and the exact point of transcriptional activation of PrP during embryogenesis is unknown. We have studied PrP mRNA expression in murine embryos and both mRNA and protein expression in a variety of adult tissues. PrP RNA was detected at different levels in all tissues tested while PrP(C) protein was detectable in all adult tissues tested with the exception of kidney and liver. RNA and protein levels were also assessed at four points during postnatal brain development and levels of both were seen to increase with development. We also established that, during embryogenesis, induction of PrP RNA expression occurs between E8.5 and E9, during the period of transition from anaerobic to aerobic metabolism. Preliminary experiments investigating the effects of superoxide radicals on PrP expression in cultured neuroblastoma and astrocyte cells support the suggestion that PrP(C) forms part of a cellular antioxidant defense mechanism.
Collapse
MESH Headings
- Aging/genetics
- Aging/metabolism
- Animals
- Animals, Newborn
- Antioxidants/metabolism
- Brain/embryology
- Brain/growth & development
- Brain/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cells, Cultured
- Embryo, Mammalian/embryology
- Embryo, Mammalian/metabolism
- Energy Metabolism/genetics
- Fetus
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Oxidative Stress/drug effects
- Oxidative Stress/genetics
- PrPC Proteins/drug effects
- PrPC Proteins/genetics
- PrPC Proteins/metabolism
- Prion Diseases/genetics
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Superoxides/pharmacology
- Transcriptional Activation/drug effects
- Transcriptional Activation/genetics
- Viscera/embryology
- Viscera/metabolism
Collapse
Affiliation(s)
- G. Miele
- *Department of Gene Expression & Development, Roslin Institute, Roslin, Midlothian, Scotland, EH25 9PS, UK
| | - A. R. Alejo Blanco
- *Department of Gene Expression & Development, Roslin Institute, Roslin, Midlothian, Scotland, EH25 9PS, UK
| | - H. Baybutt
- †BBSRC Institute for Animal Health Neuropathogenesis Unit, Ogston Building, West Mains Road, Edinburgh, Scotland, EH9 3JF, UK
| | - S. Horvat
- ‡Biotechnical Faculty, Zootechnical Department, University of Ljubljana, Slovenia
| | - J. Manson
- †BBSRC Institute for Animal Health Neuropathogenesis Unit, Ogston Building, West Mains Road, Edinburgh, Scotland, EH9 3JF, UK
| | - M. Clinton
- *Department of Gene Expression & Development, Roslin Institute, Roslin, Midlothian, Scotland, EH25 9PS, UK
- Address correspondence to M. Clinton, Department of Gene Expression & Development, Roslin Institute, Roslin, Midlothian, Scotland, EH25 9PS, UK. Tel: +44 131 527 4216; Fax: +44 131 440 0434; E-mail:
| |
Collapse
|
20
|
Salès N, Hässig R, Rodolfo K, Di Giamberardino L, Traiffort E, Ruat M, Frétier P, Moya KL. Developmental expression of the cellular prion protein in elongating axons. Eur J Neurosci 2002; 15:1163-77. [PMID: 11982627 DOI: 10.1046/j.1460-9568.2002.01953.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PrPc, a sialoglycoprotein present in the normal adult hamster brain, is particularly abundant in plastic brain regions but little is known about the level of expression and the localization of the protein during development. Western blot analysis of whole brain homogenates with mab3F4 show very low levels of the three main molecular weight forms of the protein at birth, in contrast to the strong and wide expression of mRNA transcripts. The PrPc levels increase sharply through P14 and are diminished somewhat in the adult. Regional analysis showed that in structures with ongoing growth or plasticity such as the olfactory bulb and hippocampus, PrPc remains high in the adult, while in areas where structural and functional relationships stabilize during development, such as the cortex and the thalamus, PrPc levels decline after the third postnatal week. In the neonate brain PrPc was prominent along fiber tracts similar to markers of axon elongation and in vitro experiments showed that the protein was present on the surface of elongating axons. PrPc is then localized to the synaptic neuropil in close spatio-temporal association with synapse formation. The localization of PrPc on elongating axons suggests a role for the protein in axon growth. In addition, the relative abundance of the protein in developing axon pathways and during synaptogenesis may provide a basis for the age-dependent susceptibility to transmissible spongiform encephalopathies.
Collapse
Affiliation(s)
- Nicole Salès
- INSERM U.334, Service Hospitalier Frédéric Joliot, DRM/DSV/CEA, 4 Place du Général Leclerc, 91401 Orsay Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
To develop spatio-temporally controlled somatic mutagenesis in the adult mouse nervous system, we established transgenic mice expressing the tamoxifen-inducible Cre-ERT recombinase under the control of the mouse prion protein (PrP) promoter. Cre-ERT was expressed in most regions of the brain and in the retina of one transgenic line, whereas its expression was mostly restricted to the hippocampus and the cerebellum in another line. As tamoxifen efficiently induced Cre-mediated recombination in the various neuronal cell types expressing Cre-ERT in the brain of adult mice, the PrP-Cre-ERT lines should be valuable tools to study the functions of genes involved in neurodegenerative diseases or regeneration, and in complex processes such as behaviour, learning and memory. Some limitations of presently available reporter lines for Cre-mediated recombination in adult mouse CNS are discussed.
Collapse
Affiliation(s)
- P Weber
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique, Collège de France, Université Louis Pasteur, BP163, 67404 Illkirch-Cedex, Communauté Urbaine de Strasbourg, France
| | | | | |
Collapse
|
22
|
Sun YF, Yu LY, Saarma M, Timmusk T, Arumae U. Neuron-specific Bcl-2 homology 3 domain-only splice variant of Bak is anti-apoptotic in neurons, but pro-apoptotic in non-neuronal cells. J Biol Chem 2001; 276:16240-7. [PMID: 11278671 DOI: 10.1074/jbc.m010419200] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We have identified and characterized N-Bak, a neuron-specific isoform of the pro-apoptotic Bcl-2 family member Bak. N-Bak is generated by neuron-specific splicing of a novel 20-base pair exon, which changes the previously described Bak, containing Bcl-2 homology (BH) domains BH1, BH2, and BH3, into a shorter BH3-only protein. As demonstrated by reverse transcription-polymerase chain reaction and RNase protection assay, N-Bak transcripts are expressed only in central and peripheral neurons, but not in other cells, whereas the previously described Bak is expressed ubiquitously, but not in neurons. Neonatal sympathetic neurons microinjected with N-Bak resisted apoptotic death caused by nerve growth factor (NGF) removal, whereas microinjected Bak accelerated NGF deprivation-induced death. Overexpressed Bak killed sympathetic neurons in the presence of NGF, whereas N-Bak did not. N-Bak was, however, still death-promoting when overexpressed in non-neuronal cells. Thus, N-Bak is an anti-apoptotic BH3-only protein, but only in the appropriate cellular environment. This is the first example of a neuron-specific Bcl-2 family member.
Collapse
Affiliation(s)
- Y F Sun
- Program of Molecular Neurobiology, Institute of Biotechnology, University of Helsinki, Viikki Biocenter, FIN-00014 Helsinki, Finland
| | | | | | | | | |
Collapse
|
23
|
Liu T, Zwingman T, Li R, Pan T, Wong BS, Petersen RB, Gambetti P, Herrup K, Sy MS. Differential expression of cellular prion protein in mouse brain as detected with multiple anti-PrP monoclonal antibodies. Brain Res 2001; 896:118-29. [PMID: 11277980 DOI: 10.1016/s0006-8993(01)02050-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The normal cellular prion protein (PrP(C)) plays an essential role in the development of prion diseases. Indirect evidence has suggested that different PrP(C) glycoforms may be expressed in different brain regions and perform distinct functions. However, due to a lack of monoclonal antibodies (Mabs) that are specific for mouse PrP(C), the expression of PrP(C) in the mouse brain has not been studied in great detail. We used Mabs specific for either the N-terminus or the C-terminus of the mouse PrP(C) to study its expression in the mouse brain by immunoblotting and immunohistochemistry. Immunoblotting studies demonstrated that the expression of PrP(C) differed quantitatively as well as qualitatively in different regions of the brain. The anti-C-terminus Mabs reacted with all three molecular weight bands of PrP(C); the anti-N-terminus Mabs only reacted with the 39-42 kDa PrP(C). The results from immunohistochemical staining revealed the spatial distribution of PrP(C) in the mouse brain, which were consistent with that from immunoblotting. Although expression of PrP(C) has been reported to be required for long-term survival of Purkinje cells, we were unable to detect PrP(C) in the Purkinje cell layer in the cerebellum with multiple anti-PrP Mabs. Our findings suggest that PrP(C) variants, i.e. various glycoforms and truncated forms, might be specifically expressed in different regions of mouse brain and might have different functions.
Collapse
Affiliation(s)
- T Liu
- Institute of Pathology, Case Western Reserve University, School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106-1712, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Liu T, Li R, Wong BS, Liu D, Pan T, Petersen RB, Gambetti P, Sy MS. Normal cellular prion protein is preferentially expressed on subpopulations of murine hemopoietic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:3733-42. [PMID: 11238614 DOI: 10.4049/jimmunol.166.6.3733] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We studied the expression of normal cellular prion protein (PrP(C)) in mouse lymphoid tissues with newly developed mAbs to PrP(C). Most of the mature T and B cells in the peripheral lymphoid organs do not express PrP(C). In contrast, most thymocytes are PrP(C+). In the bone marrow, erythroid cells and maturing granulocytes are PrP(C+). Approximately 50% of the cells in the region of small lymphocytes and progenitor cells also express PrP(C). Most of these PrP(C+) cells are CD43(+), but B220(-), surface IgM(-) (sIgM(-)), and IL-7R(-), a phenotype that belongs to cells not yet committed to the B cell lineage. Another small group of the PrP(C+) cell are B220(+), and some of these are also sIgM(+). The majority of the B220(+) cells, however, are PrP(C-). Therefore, PrP(C) is preferentially expressed in early bone marrow progenitor cells and subsets of maturing B cells. Supporting this interpretation is our observation that stimulation of bone marrow cells in vitro with PMA results in a decrease in the number of PrP(C+)B220(-) cells with a corresponding increase of sIgM(+)B220(high) mature B cells. This result suggests that the PrP(C+)B220(-) cells are potential progenitors. Furthermore, in the bone marrow of Rag-1(-/-) mice, there are an increased number of PrP(C+)B220(-) cells, and most of the developmentally arrested pro-B cells in these mice are PrP(C+). Collectively, these results suggest that PrP(C) is expressed preferentially in immature T cells in the thymus and early progenitor cells in the bone marrow, and the expression of PrP(C) is regulated during hemopoietic differentiation.
Collapse
Affiliation(s)
- T Liu
- Institute of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Sakaue M, Nakamura H, Kaneko I, Kawasaki Y, Arakawa N, Hashimoto H, Koyama Y, Baba A, Matsuda T. Na(+)-Ca(2+) exchanger isoforms in rat neuronal preparations: different changes in their expression during postnatal development. Brain Res 2000; 881:212-6. [PMID: 11036162 DOI: 10.1016/s0006-8993(00)02808-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We examined the relative amounts of Na(+)-Ca(2+) exchanger (NCX) isoform mRNAs in cultured neurons, astrocytes and developmental rat brain. NCX1 transcript was predominant in neurons and astrocytes, but NCX2 transcript was about four-fold higher than NCX1 or NCX3 transcript in adult rat cortex. NCX2 transcript in the cortex increased markedly during postnatal development, whereas NCX1 and NCX3 transcripts decreased. Na(+)-dependent 45Ca(2+) uptake in the cortical homogenate increased significantly during postnatal development.
Collapse
Affiliation(s)
- M Sakaue
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, 565-0871, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Teter B, Rozovsky I, Krohn K, Anderson C, Osterburg H, Finch C. Methylation of the glial fibrillary acidic protein gene shows novel biphasic changes during brain development. Glia 1996; 17:195-205. [PMID: 8840161 DOI: 10.1002/(sici)1098-1136(199607)17:3<195::aid-glia2>3.0.co;2-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The gene for glial fibrillary acidic protein (GFAP) was analyzed in the rat for developmental changes in methylation of cytosine at CpG sequences as a correlate of the onset of GFAP mRNA expression and for the effect of methylation on GFAP promoter activity. The methylation of nine CpG sites in the GFAP promoter and ten sites in exon 1 was analyzed in F344 rats by a quantitative application of ligation-mediated polymerase chain reaction. Whole rat brain poly(A)+ RNA showed an exponential increase of GFAP mRNA after embryo day 14 that reached stable adult levels by postnatal day 10. During development, only the seven CpG sites in the far-upstream promoter showed large changes in methylation; these sites constitute the brain-specific domain of methylation described in adult rats (Teter et al: J Neurosci Res 39:680, 1994). These seven CpG sites showed a cycle of demethylation during the onset of GFAP transcription in the embryo (between embryonic day 14 and postnatal day 10) followed by remethylation at later postnatal ages when GFAP mRNA remains prevalent. The minimum levels of methylation across these CpG sites displayed a gradient with the lowest minima at the 3' sites. This demethylation/remethylation cycle is a novel phenomenon in DNA methylation during perinatal development. The demethylation/remethylation cycle during development was also shown by the opposite-strand cytosines. Two cytosines in this region that are conserved in rat and mouse also undergo the same demethylation/remethylation cycle in the mouse GFAP gene during development, implying evolutionary conservation and functional significance. As a further test of functional significance, a Luciferase reporter gene assay was evaluated in primary cultured astrocytes; the activity of the GFAP promoter was reduced when it was methylated at one or all CpG sites. Therefore, the GFAP promoter may be activated in rodent development by transient demethylation of a conserved brain-specific methylation domain.
Collapse
Affiliation(s)
- B Teter
- Neurogerontology Division, Andrus Gerontology Center, University of Southern California, Los Angeles 90089-0191, USA
| | | | | | | | | | | |
Collapse
|
27
|
Yao J, Kitt C, Reeves RH. Chronic elevation of S100 beta protein does not alter APP mRNA expression or promote beta-amyloid deposition in the brains of aging transgenic mice. Brain Res 1995; 702:32-6. [PMID: 8846093 DOI: 10.1016/0006-8993(95)00991-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
S100 beta protein, a member of a Ca(2+)-binding protein family present primarily in the nervous system, affects the survival and differentiation of both neurons and glia cells. Elevated levels of S100 beta protein have been observed in the brains of individuals with Alzheimer Disease (AD), as well as in those with Down Syndrome (DS). We have examined transcript levels from the gene encoding the amyloid precursor protein (APP) in four brain regions of mice from 1 to 24 months of age. After stable adult levels of expression are reached, APP mRNA levels do not change with aging. APP mRNA levels are independent of normal regional variation in S100 beta mRNA and protein. Further, chronic exposure to S100 beta elevated 2- or 7-fold above normal did not alter the transcript levels of APP in transgenic mice. These results leave open the possibility of focal changes in APP transcription and do not address possible effects of S100 beta on the complex processing known to occur with APP protein. However, neither control nor transgenic aged mice showed any evidence of abnormal deposition of amyloid in neuritic plaques. These results are discussed in the context of hypotheses about the role of elevated S100 beta in DS and AD.
Collapse
Affiliation(s)
- J Yao
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
28
|
Chen SG, Teplow DB, Parchi P, Teller JK, Gambetti P, Autilio-Gambetti L. Truncated forms of the human prion protein in normal brain and in prion diseases. J Biol Chem 1995; 270:19173-80. [PMID: 7642585 DOI: 10.1074/jbc.270.32.19173] [Citation(s) in RCA: 387] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The cellular form of the prion protein (PrPc) is a glycoprotein anchored to the cell membrane by a glycosylphosphatidylinositol moiety. An aberrant form of PrPc that is partially resistant to proteases, PrPres, is a hallmark of prion diseases, which in humans include Cruetzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome, and fatal familial insomnia. We have characterized the major forms of PrP in normal and pathological human brains. A COOH-terminal fragment of PrPc, designated C1, is abundant in normal and CJD brains as well as in human neuroblastoma cells. Sequence analysis revealed that C1 contains alternative NH2 termini starting at His-111 or Met-112. Like PrPc, C1 is glycosylated, anchored to the cell membrane, and is heat-stable. Consistent with the lack of the NH2-terminal region of PrPc, C1 is more acidic than PrPc and does not bind heparin. An additional fragment longer than C1, designated C2, is present in substantial amounts in CJD brains. Like PrPres, C2 is resistant to proteases and is detergent-insoluble. Our data indicate that C1 is a major product of normal PrPc metabolism, generated by a cleavage that disrupts the neurotoxic and amyloidogenic region of PrP comprising residues 106-126. This region remains intact in C2, suggesting a role for C2 in prion diseases.
Collapse
Affiliation(s)
- S G Chen
- Division of Neuropathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | |
Collapse
|
29
|
Schuller AG, Groffen C, van Neck JW, Zwarthoff EC, Drop SL. cDNA cloning and mRNA expression of the six mouse insulin-like growth factor binding proteins. Mol Cell Endocrinol 1994; 104:57-66. [PMID: 7529732 DOI: 10.1016/0303-7207(94)90051-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The insulin-like growth factor binding proteins (IGFBPs) comprise a family of six distinct proteins which modulate insulin-like growth factor action. We have isolated cDNAs encoding the six mouse IGFBPs (mIGFBPs). In addition, we studied the mRNA expression of the six mIGFBPs during development and in various adult tissues. Our results show that each of the six mIGFBPs is highly homologous to their human and rat counterparts, whereas only the N and C terminal ends are conserved between the six mIGFBPs. Northern blotting revealed that mIGFBP-2, -3, -4 and -5 genes are already expressed at gestational day 11.5, suggesting a role for these mIGFBPs in embryonal development. In liver, a peak of mIGFBP-1 mRNA expression was found around birth, suggesting a function for mIGFBP-1 in the newborn mouse. Finally, tissue-specific expression of the six mouse IGFBP genes was observed in adult tissues suggesting different roles or modes of actions in adult life.
Collapse
Affiliation(s)
- A G Schuller
- Department of Pediatrics, Erasmus University/Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
30
|
Lazarini F, Boussin F, Deslys JP, Tardy M, Dormont D. Astrocyte gene expression in experimental mouse scrapie. J Comp Pathol 1994; 111:87-98. [PMID: 7962730 DOI: 10.1016/s0021-9975(05)80114-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The biological hallmark of transmissible spongiform encephalopathies is a significant accumulation, in brain, of the scrapie prion protein (PrPsc), often associated with an increased glial fibrillary acidic protein (GFAP) expression. This study was focused on astrocyte gene expression during scrapie development over a period of 172 days in intracerebrally inoculated newborn mice. The levels of expression of PrP and two specific astrocyte proteins, -GFAP and glutamine synthetase (GS)-, were investigated by Western and Northern blots. In brain, a 10-fold increased expression of GFAP mRNAS was demonstrated from 112 days post-inoculation to 172 days, whereas the "upregulation" of GS mRNAs was two-fold. GFAP was observed to increase 10- to 20-fold in scrapie-infected brain from day 112 to day 172, while PrP showed a three- to four-fold elevation. Both proteins were found in greater amount in the frontal cortex and cerebellum of animals with clinical scrapie than in those given an injection of normal brain. PrPsc was detected in scrapie brain from day 84 after inoculation, and thereafter increased about 20-fold until day 172. On the other hand, the concentration of glutamine synthetase remained constant in brain throughout the scrapie disease. To conclude, these results show that GFAP and GS mRNAs are differently upregulated in brain in the scrapie mouse model.
Collapse
Affiliation(s)
- F Lazarini
- Laboratoire de Neuropathologie Expérimentale et Neurovirologie, CRSSA, Commissariat à l'Energie Atomique, DPTE/DSV, Fontenay aux Roses, France
| | | | | | | | | |
Collapse
|
31
|
Voronina AS, Preobrazhensky AA. Developmental expression of glial fibrillary acidic protein gene in human embryos. Neurosci Lett 1994; 174:198-200. [PMID: 7970178 DOI: 10.1016/0304-3940(94)90020-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Glial fibrillary acidic protein (GFAP) is an intermediate filament protein of astrocytes. We have shown by Northern blot analysis that GFAP mRNA first appears in the spinal cord at the age of 8 weeks and in the brain at the age of 9 weeks of embryogenesis, and its relative contents increases at later stages. A plasmid selected from a cDNA expression library with an insert encoding for almost full length of human GFAP was used in this study.
Collapse
Affiliation(s)
- A S Voronina
- A.N. Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow
| | | |
Collapse
|
32
|
Lazarini F, Castelnau P, Chermann JF, Deslys JP, Dormont D. Modulation of prion protein gene expression by growth factors in cultured mouse astrocytes and PC-12 cells. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1994; 22:268-74. [PMID: 7912403 DOI: 10.1016/0169-328x(94)90055-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The present study was performed on primary cultures of mouse astrocytes and cultures of rat pheochromocytoma PC-12 in order to investigate the regulation of the prion protein (PrP) gene expression in relation to proliferation and differentiation. Treatment of PC-12 cells with interleukin-6 (IL-6) and beta-nerve growth factor (NGF) resulted in induction of neuronal differentiation. Northern blot analysis demonstrated a 4-fold increase of PrP mRNA in relation to cellular differentiation, after 7 days of treatment with either of the two factors. In astrocytes, PrP and glial fibrillary acidic protein (GFAP) mRNA levels were found to be regulated in a similar manner during development in vitro. A 3-fold increase of their mRNAs was observed from 5 to 14 days of culture (proliferation period). Then, their gene expressions showed a slight decrease from 14 to 28 days (maturation period). Treatment of astrocytes with IL-6, basic fibroblast growth factor (bFGF), and epidermal growth factor (EGF) appeared to markedly down-regulate the expression of GFAP mRNAs, which might reflect cell maturation. In contrast, they had no significant effect on the expression of PrP gene. These results suggest that the PrP gene expression is differently regulated in neural cells. In neuronal cells, it is mainly associated with differentiation. On the other hand, in astrocytes, the PrP mRNA level seems to be not only related to the proliferation and differentiation stages.
Collapse
Affiliation(s)
- F Lazarini
- Laboratoire de Neuropathologie Expérimentale et Neurovirologie, CRSSA, Commissariat à l'Energie Atomique, DPTE/DSV, Fontenay-aux Roses, France
| | | | | | | | | |
Collapse
|
33
|
Ekblom J, Jossan SS, Bergström M, Oreland L, Walum E, Aquilonius SM. Monoamine oxidase-B in astrocytes. Glia 1993; 8:122-32. [PMID: 8406673 DOI: 10.1002/glia.440080208] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In the present report we describe the astrocytic localization and content of monoamine oxidase-B (MAO-B) by means of a 3H-L-deprenyl emulsion autoradiography in primary cultures of rat astrocytes, in cryosectioned astrocytoma surgical specimen, and in cryosections of human spinal cords from patients dying in amyotrophic lateral sclerosis (ALS) and controls. The occurrence of MAO-B enzyme protein depends on the degree of cellular differentiation as demonstrated by studies on astrocytes in primary cultures analyzed at two different stages of maturation. Highly differentiated cells exhibited high relative enzyme concentration whereas glioblasts lacked or showed very low contents of MAO-B enzyme. This was further substantiated by studies performed on human astrocytoma tissue using 3H-L-deprenyl emulsion autoradiography in combination with immunohistochemical detection of glial fibrillary acidic protein (GFAP). Regional increases of MAO-B concentration were found in ALS lumbar sections with quantitative 3H-L-deprenyl autoradiography. On the basis of results obtained from double staining for GFAP and MAO-B, the increase in MAO-B seemed to be due to an increased number of astrocytes as well as an increased content of MAO-B in reactive species of astrocytes. A cell culture model has been used that produces cells with morphology and GFAP-content similar to reactive cells. These astrocytes exhibited high relative content of the MAO-B enzyme protein. In the light of the presented data, taking into account the finding that a subpopulation of reactive cells contained low levels of MAO-B, a heterogeneity among reactive astrocytes was observed.
Collapse
Affiliation(s)
- J Ekblom
- Department of Neurology, University Hospital, Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
34
|
Harris DA, Lele P, Snider WD. Localization of the mRNA for a chicken prion protein by in situ hybridization. Proc Natl Acad Sci U S A 1993; 90:4309-13. [PMID: 8483948 PMCID: PMC46496 DOI: 10.1073/pnas.90.9.4309] [Citation(s) in RCA: 60] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The infectious agent (prion) responsible for transmissible spongiform encephalopathies in humans and animals is composed primarily of a 33- to 35-kDa glycoprotein called PrPSc (scrapie isoform of prion protein), which is a posttranslationally modified form of the normal cell-surface protein PrPC. Little is known about the function of PrPC. Interestingly, chPrP, the chicken homologue of PrPC, copurifies with a factor from brain that stimulates synthesis of acetylcholine receptors on skeletal muscle cells. Using in situ hybridization, we report here that chPrP mRNA is widely distributed in cholinergic and noncholinergic neurons throughout the adult central nervous system, including those in the telencephalic striata, thalamus and hypothalamus, optic tectum, medulla, cerebellum, and spinal cord. The mRNA is present in the brain and spinal cord as early as embryonic day 6 and is also found in dorsal root ganglia, retina, intestine, and heart. Our data suggest that if chPrP serves to regulate acetylcholine receptor number on postsynaptic targets, this is not its only function. It is likely that the protein plays a more widespread role in the central nervous system and perhaps elsewhere, possibly one related to intercellular communication, adhesion, or recognition. The chicken embryo represents an attractive experimental system in which to investigate the normal developmental function of PrPC.
Collapse
Affiliation(s)
- D A Harris
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
| | | | | |
Collapse
|
35
|
Burns TM, Clough JA, Klein RM, Wood GW, Berman NE. Developmental regulation of cytokine expression in the mouse brain. Growth Factors 1993; 9:253-8. [PMID: 8148154 DOI: 10.3109/08977199308991585] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Expression of four cytokine genes, transforming growth factor (TGF) beta 2, tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and macrophage colony-stimulating factor (CSF-1 also known as M-CSF) was examined to determine whether these genes are developmentally regulated in the brain. Northern blots were performed on RNA isolated from the mouse brain from embryonic day 15 (E15) through postnatal day 9. TGF beta 2 gene expression was relatively high in the earliest embryos studied and decreased after E16-E17, and the three transcripts were developmentally regulated. TNF-alpha and IL-6 were detected in total RNA on all days studied. CSF-1 was detected only in polyadenylated RNA. The data suggest that expression of these cytokines is related to specific developmental events that share cellular functions with regenerative or inflammatory processes.
Collapse
Affiliation(s)
- T M Burns
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City 66160-7400
| | | | | | | | | |
Collapse
|
36
|
Abstract
This review summarized a part of our studies over a long period of time, relating them to the literature on the same topics. We aimed our research toward an understanding of the genetic origin of brain specific proteins, identified by B. W. Moore and of the high complexity of the nucleotide sequence of brain mRNA, originally investigated by W. E. Hahn, but have not completely achieved the projected goal. According to our studies, the reason for the high complexity in the RNA of brain nuclei might be the high complexity in neuronal nuclear RNA as described in the Introduction. Although one possible explanation is that it results from the summation of RNA complexities of several neuronal types, our saturation hybridization study with RNA from the isolated nuclei of granule cells showed an equally high sequence complexity as that of brain. It is likely that this type of neuron also contains numerous rare proteins and peptides, perhaps as many as 20,000 species which were not detectable even by two-dimensional PAGE. I was possible to gain insight into the reasons for the high sequence complexity of brain RNA by cloning the cDNA and genomic DNA of the brain-specific proteins as described in the previous sections. These data provided evidence for the long 3'-noncoding regions in the cDNA of the brain-specific proteins which caused the mRNA of brain to be larger than that from other tissues. During isolation of such large mRNAs, a molecule might be split into a 3'-poly(A)+RNA and 5'-poly(A)-RNA. In the studies on genomic DNA, genes with multiple transcription initiation sites were found in brain, such as CCK, CNP and MAG, in addition to NSE which was a housekeeping gene, and this may contribute to the high sequence complexity of brain RNA. Our studies also indicated the presence of genes with alternative splicing in brain, such as those for CNP, MAG and NGF, suggesting a further basis for greater RNA nucleotide sequence complexity. It is noteworthy that alternative splicing of the genes for MBP and PLP also produced multiple mRNAs. Such a mechanism may be a general characteristic of the genes for the myelin-specific proteins produced by oligodendrocytes. In considering the high nucleotide sequence complexity, it is interesting that MAG and S-100 beta genes etc. possess two additional sites for poly(A).(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- Y Takahashi
- Department of Neuropharmacology, Niigata University, Japan
| |
Collapse
|
37
|
Sakata M, Farooqui SM, Prasad C. Post-transcriptional regulation of loss of rat striatal D2 dopamine receptor during aging. Brain Res 1992; 575:309-14. [PMID: 1533340 DOI: 10.1016/0006-8993(92)90095-q] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The mechanism(s) underlying age-associated diminutions in the rat striatal D2 dopamine receptor (D2-receptor) number was investigated. The levels of D2-receptor mRNA in 4-, 12- and 18-month-old rat striata were found not to change. In contrast, the levels of 110 kDa protein, labeled with a D2-receptor specific antibody, decreased in parallel with [3H]YM-09151-2 binding to striatal membranes. These data suggest a role for post-transcriptional mechanism(s) in age-associated decrease in D2-receptor.
Collapse
Affiliation(s)
- M Sakata
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge 70808
| | | | | |
Collapse
|
38
|
Xu SX, Monsma FJ, Sibley DR, Creese I. Regulation of D1A and D2 dopamine receptor mRNA during ontogenesis, lesion and chronic antagonist treatment. Life Sci 1992; 50:383-96. [PMID: 1732708 DOI: 10.1016/0024-3205(92)90440-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The developmental characteristics of D1A and D2 dopamine receptor mRNA levels were determined by Northern blot analyses. Striatal D1A and D2 dopamine receptor mRNAs of male Fischer 344 rats were about 60% of adult (day 120) levels at postnatal day 1 and reached their highest levels at day 30 (126 and 139% adult levels) and then decreased by day 120 (100%). D1 and D2 dopamine receptors showed much greater quantitative changes with densities at day 30 about 6- and 14-fold higher than at day 1, respectively, while mRNA levels showed only a 2-fold increase. The highest level of D2 dopamine receptor mRNA in the midbrain was reached at day 14 (195% of adult levels) while the level at day 1 was 31% higher than that at day 120. Striatal beta-actin mRNA levels decreased gradually as the rats developed with the level at postnatal day 1 almost twice that at day 120 postpartum. Treatment of adult rats with the selective D2 dopamine receptor antagonist, haloperidol (0.5 mg/kg/day, s.c., for 2 h, 7, 14, 21 days or 21 days + 3 days withdrawal) had no effect on striatal D2 dopamine receptor mRNA levels in spite of significant increases in dopamine receptor density at the later time points. However, 21 days following a 6-hydroxydopamine lesion of the nigrostriatal pathway, striatal D2 dopamine receptor mRNA levels were increased by 53%.
Collapse
Affiliation(s)
- S X Xu
- Center for Molecular and Behavioral Neuroscience, Rutgers, State University of New Jersey, Newark 07102
| | | | | | | |
Collapse
|