Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.676
Filter
1.
Cell ; 171(1): 120-132.e12, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28919079

ABSTRACT

The disassembly of the intron lariat spliceosome (ILS) marks the end of a splicing cycle. Here we report a cryoelectron microscopy structure of the ILS complex from Saccharomyces cerevisiae at an average resolution of 3.5 Å. The intron lariat remains bound in the spliceosome whereas the ligated exon is already dissociated. The step II splicing factors Prp17 and Prp18, along with Cwc21 and Cwc22 that stabilize the 5' exon binding to loop I of U5 small nuclear RNA (snRNA), have been released from the active site assembly. The DEAH family ATPase/helicase Prp43 binds Syf1 at the periphery of the spliceosome, with its RNA-binding site close to the 3' end of U6 snRNA. The C-terminal domain of Ntr1/Spp382 associates with the GTPase Snu114, and Ntr2 is anchored to Prp8 while interacting with the superhelical domain of Ntr1. These structural features suggest a plausible mechanism for the disassembly of the ILS complex.


Subject(s)
Introns , Spliceosomes/ultrastructure , Cryoelectron Microscopy , DEAD-box RNA Helicases/chemistry , Models, Molecular , RNA Precursors/chemistry , RNA Precursors/ultrastructure , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/ultrastructure , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Schizosaccharomyces/chemistry , Spliceosomes/chemistry
2.
Cell ; 171(7): 1589-1598.e8, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29153833

ABSTRACT

Removal of an intron from a pre-mRNA by the spliceosome results in the ligation of two exons in the post-catalytic spliceosome (known as the P complex). Here, we present a cryo-EM structure of the P complex from Saccharomyces cerevisiae at an average resolution of 3.6 Å. The ligated exon is held in the active site through RNA-RNA contacts. Three bases at the 3' end of the 5' exon remain anchored to loop I of U5 small nuclear RNA, and the conserved AG nucleotides of the 3'-splice site (3'SS) are specifically recognized by the invariant adenine of the branch point sequence, the guanine base at the 5' end of the 5'SS, and an adenine base of U6 snRNA. The 3'SS is stabilized through an interaction with the 1585-loop of Prp8. The P complex structure provides a view on splice junction formation critical for understanding the complete splicing cycle.


Subject(s)
Saccharomyces cerevisiae/chemistry , Spliceosomes/chemistry , Cryoelectron Microscopy , Humans , Models, Molecular , RNA Splicing , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism
3.
Mol Cell ; 82(15): 2769-2778.e4, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35705093

ABSTRACT

Pre-mRNA splicing involves two sequential reactions: branching and exon ligation. The C complex after branching undergoes remodeling to become the C∗ complex, which executes exon ligation. Here, we report cryo-EM structures of two intermediate human spliceosomal complexes, pre-C∗-I and pre-C∗-II, both at 3.6 Å. In both structures, the 3' splice site is already docked into the active site, the ensuing 3' exon sequences are anchored on PRP8, and the step II factor FAM192A contacts the duplex between U2 snRNA and the branch site. In the transition of pre-C∗-I to pre-C∗-II, the step II factors Cactin, FAM32A, PRKRIP1, and SLU7 are recruited. Notably, the RNA helicase PRP22 is positioned quite differently in the pre-C∗-I, pre-C∗-II, and C∗ complexes, suggesting a role in 3' exon binding and proofreading. Together with information on human C and C∗ complexes, our studies recapitulate a molecular choreography of the C-to-C∗ transition, revealing mechanistic insights into exon ligation.


Subject(s)
Saccharomyces cerevisiae Proteins , Spliceosomes , Exons/genetics , Humans , RNA Precursors/metabolism , RNA Splice Sites , RNA Splicing , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA, Small Nuclear/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism
4.
Mol Cell ; 75(3): 538-548.e3, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31229405

ABSTRACT

The RNA catalytic core of spliceosomes as visualized by cryoelectron microscopy (cryo-EM) remains unchanged at different stages of splicing. However, we demonstrate that mutations within the core of yeast U6 snRNA modulate conformational changes between the two catalytic steps. We propose that the intramolecular stem-loop (ISL) of U6 exists in two competing states, changing between a default, non-catalytic conformation and a transient, catalytic conformation. Whereas stable interactions in the catalytic triplex promote catalysis and their disruptions favor exit from the catalytic conformation, destabilization of the lower ISL stem promotes catalysis and its stabilization supports exit from the catalytic conformation. Thus, in addition to the catalytic triplex, U6-ISL acts as an important dynamic component of the catalytic center. The relative flexibility of the lower U6-ISL stem is conserved across eukaryotes. Similar features are found in U6atac and domain V of group II introns, arguing for the generality of the proposed mechanism.


Subject(s)
Alternative Splicing/genetics , RNA, Small Nuclear/ultrastructure , Ribonucleoprotein, U4-U6 Small Nuclear/ultrastructure , Spliceosomes/ultrastructure , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Catalysis , Cryoelectron Microscopy , Introns/genetics , Mutation/genetics , Nucleic Acid Conformation , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Spliceosomes/chemistry , Spliceosomes/genetics
5.
Genes Dev ; 33(21-22): 1555-1574, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31558568

ABSTRACT

The termination of pre-mRNA splicing functions to discard suboptimal substrates, thereby enhancing fidelity, and to release excised introns in a manner coupled to spliceosome disassembly, thereby allowing recycling. The mechanism of termination, including the RNA target of the DEAH-box ATPase Prp43p, remains ambiguous. We discovered a critical role for nucleotides at the 3' end of the catalytic U6 small nuclear RNA in splicing termination. Although conserved sequence at the 3' end is not required, 2' hydroxyls are, paralleling requirements for Prp43p biochemical activities. Although the 3' end of U6 is not required for recruiting Prp43p to the spliceosome, the 3' end cross-links directly to Prp43p in an RNA-dependent manner. Our data indicate a mechanism of splicing termination in which Prp43p translocates along U6 from the 3' end to disassemble the spliceosome and thereby release suboptimal substrates or excised introns. This mechanism reveals that the spliceosome becomes primed for termination at the same stage it becomes activated for catalysis, implying a requirement for stringent control of spliceosome activity within the cell.


Subject(s)
Adenosine Triphosphatases/metabolism , DEAD-box RNA Helicases/metabolism , RNA Splicing/physiology , RNA, Small Nuclear/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism , Introns/genetics , Protein Binding , RNA Splicing/genetics
6.
RNA ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38876504

ABSTRACT

The spliceosome performs two consecutive transesterification reactions using one catalytic center, thus requiring its rearrangement between the two catalytic steps of splicing. The Prp16 ATPase facilitates exit from the first-step conformation of the catalytic center by destabilizing some interactions important for catalysis. To better understand rearrangements within the S. cerevisiae catalytic center, we characterize factors that modulate function of Prp16: Cwc2, N-terminal domain of Prp8, and U6-41AACAAU46 region. Alleles of these factors were identified through genetic screens for mutants that correct cs defects of prp16-302 allele. Several of the identified U6, cwc2, and prp8 alleles are located in close proximity of each other in cryo-EM structures of the spliceosomal catalytic conformations. Cwc2 and U6 interact with the intron sequences in the first step, but they do not seem to contribute to the stability of the second step catalytic center. On the other hand, the N-terminal segment of Prp8 not only affects intron positioning for the first step, but it also makes important contacts in the proximity of the active site for both the first and the second steps of splicing. By identifying interactions important for the stability of catalytic conformations, our genetic analyses indirectly inform us about features of the transition-state conformation of the spliceosome.

7.
RNA ; 30(4): 404-417, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38282418

ABSTRACT

RNA helicases drive necessary rearrangements and ensure fidelity during the pre-mRNA splicing cycle. DEAD-box helicase DDX41 has been linked to human disease and has recently been shown to interact with DEAH-box helicase PRP22 in the spliceosomal C* complex, yet its function in splicing remains unknown. Depletion of DDX41 homolog SACY-1 from somatic cells has been previously shown to lead to changes in alternative 3' splice site (3'ss) usage. Here, we show by transcriptomic analysis of published and novel data sets that SACY-1 perturbation causes a previously unreported pattern in alternative 3' splicing in introns with pairs of 3' splice sites ≤18 nt away from each other. We find that both SACY-1 depletion and the allele sacy-1(G533R) lead to a striking unidirectional increase in the usage of the proximal (upstream) 3'ss. We previously discovered a similar alternative splicing pattern between germline tissue and somatic tissue, in which there is a unidirectional increase in proximal 3'ss usage in the germline for ∼200 events; many of the somatic SACY-1 alternative 3' splicing events overlap with these developmentally regulated events. We generated targeted mutant alleles of the Caenorhabditis elegans homolog of PRP22, mog-5, in the region of MOG-5 that is predicted to interact with SACY-1 based on the human C* structure. These viable alleles, and a mimic of the myelodysplastic syndrome-associated allele DDX41(R525H), all promote the usage of proximal alternative adjacent 3' splice sites. We show that PRP22/MOG-5 and DDX41/SACY-1 have overlapping roles in proofreading the 3'ss.


Subject(s)
RNA Splice Sites , Spliceosomes , Humans , RNA Splice Sites/genetics , Spliceosomes/genetics , Spliceosomes/metabolism , RNA Splicing , Alternative Splicing , RNA Helicases/genetics , RNA Helicases/metabolism , DNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism
8.
RNA ; 30(7): 854-865, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38627018

ABSTRACT

The Prp19 complex (Prp19C), also named NineTeen Complex (NTC), is conserved from yeast to human and functions in many different processes such as genome stability, splicing, and transcription elongation. In the latter, Prp19C ensures TREX occupancy at transcribed genes. TREX, in turn, couples transcription to nuclear mRNA export by recruiting the mRNA exporter to transcribed genes and consequently to nascent mRNAs. Here, we assess the function of the nonessential Prp19C subunit Syf2 and the nonessential Prp19C-associated protein Cwc15 in the interaction of Prp19C and TREX with the transcription machinery, Prp19C and TREX occupancy, and transcription elongation. Whereas both proteins are important for Prp19C-TREX interaction, Syf2 is needed for full Prp19C occupancy, and Cwc15 is important for the interaction of Prp19C with RNA polymerase II and TREX occupancy. These partially overlapping functions are corroborated by a genetic interaction between Δcwc15 and Δsyf2 Finally, Cwc15 also interacts genetically with the transcription elongation factor Dst1 and functions in transcription elongation. In summary, we uncover novel roles of the Prp19C component Syf2 and the Prp19C-associated protein Cwc15 in Prp19C's function in transcription elongation.


Subject(s)
RNA Polymerase II , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Transcription Elongation, Genetic , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Protein Binding , Transcription Factors/metabolism , Transcription Factors/genetics , RNA Splicing Factors
9.
Mol Cell ; 69(6): 979-992.e6, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29547724

ABSTRACT

Human nineteen complex (NTC) acts as a multimeric E3 ubiquitin ligase in DNA repair and splicing. The transfer of ubiquitin is mediated by Prp19-a homotetrameric component of NTC whose elongated coiled coils serve as an assembly axis for two other proteins called SPF27 and CDC5L. We find that Prp19 is inactive on its own and have elucidated the structural basis of its autoinhibition by crystallography and mutational analysis. Formation of the NTC core by stepwise assembly of SPF27, CDC5L, and PLRG1 onto the Prp19 tetramer enables ubiquitin ligation. Protein-protein crosslinking of NTC, functional assays in vitro, and assessment of its role in DNA damage response provide mechanistic insight into the organization of the NTC core and the communication between PLRG1 and Prp19 that enables E3 activity. This reveals a unique mode of regulation for a complex E3 ligase and advances understanding of its dynamics in various cellular pathways.


Subject(s)
DNA Repair Enzymes/metabolism , Nuclear Proteins/metabolism , RNA Splicing Factors/metabolism , Animals , Cell Cycle Proteins/metabolism , Crystallization , DNA Damage , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Mutation , Neoplasm Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Conformation , RNA Splicing Factors/chemistry , RNA Splicing Factors/genetics , RNA-Binding Proteins/metabolism , Replication Protein A/metabolism , Sf9 Cells , Spodoptera , Structure-Activity Relationship , Ubiquitination , WD40 Repeats
10.
Mol Cell ; 67(3): 423-432.e4, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28712727

ABSTRACT

Accurate pre-mRNA splicing is needed for correct gene expression and relies on faithful splice site recognition. Here, we show that the ubiquitin-like protein Hub1 binds to the DEAD-box helicase Prp5, a key regulator of early spliceosome assembly, and stimulates its ATPase activity thereby enhancing splicing and relaxing fidelity. High Hub1 levels enhance splicing efficiency but also cause missplicing by tolerating suboptimal splice sites and branchpoint sequences. Notably, Prp5 itself is regulated by a Hub1-dependent negative feedback loop. Since Hub1-mediated splicing activation induces cryptic splicing of Prp5, it also represses Prp5 protein levels and thus curbs excessive missplicing. Our findings indicate that Hub1 mediates enhanced, but error-prone splicing, a mechanism that is tightly controlled by a feedback loop of PRP5 cryptic splicing activation.


Subject(s)
Ligases/metabolism , RNA Precursors/metabolism , RNA Splice Sites , RNA Splicing , RNA, Fungal/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Spliceosomes/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Feedback, Physiological , Gene Expression Regulation, Fungal , Hydrolysis , Ligases/chemistry , Ligases/genetics , Models, Molecular , Mutation , Protein Binding , Protein Conformation , RNA Precursors/genetics , RNA, Fungal/genetics , RNA, Messenger/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Spliceosomes/genetics , Structure-Activity Relationship , Time Factors
11.
Genes Dev ; 31(23-24): 2416-2429, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29330354

ABSTRACT

The precise function of the trimeric retention and splicing (RES) complex in pre-mRNA splicing remains unclear. Here we dissected the role of RES during the assembly and activation of yeast spliceosomes. The efficiency of pre-mRNA splicing was significantly lower in the absence of the RES protein Snu17, and the recruitment of its binding partners, Pml1 (pre-mRNA leakage protein 1) and Bud13 (bud site selection protein 13), to the spliceosome was either abolished or substantially reduced. RES was not required for the assembly of spliceosomal B complexes, but its absence hindered efficient Bact complex formation. ΔRES spliceosomes were no longer strictly dependent on Prp2 activity for their catalytic activation, suggesting that they are structurally compromised. Addition of Prp2, Spp2, and UTP to affinity-purified ΔRES B or a mixture of B/Bact complexes formed on wild-type pre-mRNA led to their disassembly. However, no substantial disassembly was observed with ΔRES spliceosomes formed on a truncated pre-mRNA that allows Prp2 binding but blocks its activity. Thus, in the absence of RES, Prp2 appears to bind prematurely, leading to the disassembly of the ΔRES B complexes to which it binds. Our data suggest that Prp2 can dismantle B complexes with an aberrant protein composition, suggesting that it may proofread the spliceosome's RNP structure prior to activation.


Subject(s)
RNA Splicing/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Spliceosomes/metabolism , DEAD-box RNA Helicases/metabolism , Protein Multimerization/genetics , RNA Precursors/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Spliceosomes/genetics
12.
Genes Dev ; 31(7): 688-701, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28446597

ABSTRACT

Multiple lines of evidence implicate chromatin in the regulation of premessenger RNA (pre-mRNA) splicing. However, the influence of chromatin factors on cotranscriptional splice site usage remains unclear. Here we investigated the function of the highly conserved histone variant H2A.Z in pre-mRNA splicing using the intron-rich model yeast Schizosaccharomyces pombe Using epistatic miniarray profiles (EMAPs) to survey the genetic interaction landscape of the Swr1 nucleosome remodeling complex, which deposits H2A.Z, we uncovered evidence for functional interactions with components of the spliceosome. In support of these genetic connections, splicing-specific microarrays show that H2A.Z and the Swr1 ATPase are required during temperature stress for the efficient splicing of a subset of introns. Notably, affected introns are enriched for H2A.Z occupancy and more likely to contain nonconsensus splice sites. To test the significance of the latter correlation, we mutated the splice sites in an affected intron to consensus and found that this suppressed the requirement for H2A.Z in splicing of that intron. These data suggest that H2A.Z occupancy promotes cotranscriptional splicing of suboptimal introns that may otherwise be discarded via proofreading ATPases. Consistent with this model, we show that overexpression of splicing ATPase Prp16 suppresses both the growth and splicing defects seen in the absence of H2A.Z.


Subject(s)
Histones/genetics , Introns , RNA Splicing , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces/genetics , Adenosine Triphosphatases/metabolism , Gene Expression Regulation, Fungal , Nucleosomes/genetics , Promoter Regions, Genetic , Schizosaccharomyces/growth & development , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Spliceosomes/genetics
13.
J Biol Chem ; 299(8): 105054, 2023 08.
Article in English | MEDLINE | ID: mdl-37454740

ABSTRACT

Neurodegenerative diseases are often characterized by the codeposition of different amyloidogenic proteins, normally defining distinct proteinopathies. An example is represented by prion diseases, where the classical deposition of the aberrant conformational isoform of the prion protein (PrPSc) can be associated with tau insoluble species, which are usually involved in another class of diseases called tauopathies. How this copresence of amyloidogenic proteins can influence the progression of prion diseases is still a matter of debate. Recently, the cellular form of the prion protein, PrPC, has been investigated as a possible receptor of amyloidogenic proteins, since its binding activity with Aß, tau, and α-synuclein has been reported, and it has been linked to several neurotoxic behaviors exerted by these proteins. We have previously shown that the treatment of chronically prion-infected cells with tau K18 fibrils reduced PrPSc levels. In this work, we further explored this mechanism by using another tau construct that includes the sequence that forms the core of Alzheimer's disease tau filaments in vivo to obtain a distinct fibril type. Despite a difference of six amino acids, these two constructs form fibrils characterized by distinct biochemical and biological features. However, their effects on PrPSc reduction were comparable and probably based on the binding to PrPC at the plasma membrane, inhibiting the pathological conversion event. Our results suggest PrPC as receptor for different types of tau fibrils and point out a role of tau amyloid fibrils in preventing the pathological PrPC to PrPSc conformational change.


Subject(s)
Neurodegenerative Diseases , Prion Diseases , Prions , tau Proteins , Humans , Amyloidogenic Proteins , Prion Diseases/metabolism , Prion Proteins , Prions/metabolism , tau Proteins/metabolism
14.
J Biol Chem ; 299(11): 105319, 2023 11.
Article in English | MEDLINE | ID: mdl-37802314

ABSTRACT

Mis-folding of the prion protein (PrP) is known to cause neurodegenerative disease; however, the native function of this protein remains poorly defined. PrP has been linked with many cellular functions, including cellular proliferation and senescence. It is also known to influence epidermal growth factor receptor (EGFR) signaling, a pathway that is itself linked with both cell growth and senescence. Adult neural stem cells (NSCs) persist at low levels in the brain throughout life and retain the ability to proliferate and differentiate into new neural lineage cells. KO of PrP has previously been shown to reduce NSC proliferative capacity. We used PrP KO and WT NSCs from adult mouse brain to examine the influence of PrP on cellular senescence, EGFR signaling, and the downstream cellular processes. PrP KO NSCs showed decreased cell proliferation and increased senescence in in vitro cultures. Expression of EGFR was decreased in PrP KO NSCs compared with WT NSCs and additional supplementation of EGF was sufficient to reduce senescence. RNA-seq analysis confirmed that significant changes were occurring at the mRNA level within the EGFR signaling pathway and these were associated with reduced expression of mitochondrial components and correspondingly reduced mitochondrial function. Metabolomic analysis of cellular energy pathways showed that blockages were occurring at critical sites for production of energy and biomass, including catabolism of pyruvate. We conclude that, in the absence of PrP, NSC growth pathways are downregulated as a consequence of insufficient energy and growth intermediates.


Subject(s)
Neural Stem Cells , Neurodegenerative Diseases , Prions , Animals , Mice , Cell Proliferation , Cellular Senescence , ErbB Receptors/genetics , ErbB Receptors/metabolism , Neural Stem Cells/metabolism , Neurodegenerative Diseases/metabolism , Prion Proteins/genetics , Prion Proteins/metabolism , Prions/metabolism , Signal Transduction/genetics , Mice, Inbred C57BL
15.
Emerg Infect Dis ; 30(6): 1193-1202, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38781931

ABSTRACT

Chronic wasting disease (CWD) is a cervid prion disease with unknown zoonotic potential that might pose a risk to humans who are exposed. To assess the potential of CWD to infect human neural tissue, we used human cerebral organoids with 2 different prion genotypes, 1 of which has previously been associated with susceptibility to zoonotic prion disease. We exposed organoids from both genotypes to high concentrations of CWD inocula from 3 different sources for 7 days, then screened for infection periodically for up to 180 days. No de novo CWD propagation or deposition of protease-resistant forms of human prions was evident in CWD-exposed organoids. Some persistence of the original inoculum was detected, which was equivalent in prion gene knockout organoids and thus not attributable to human prion propagation. Overall, the unsuccessful propagation of CWD in cerebral organoids supports a strong species barrier to transmission of CWD prions to humans.


Subject(s)
Organoids , Prions , Wasting Disease, Chronic , Wasting Disease, Chronic/transmission , Humans , Prions/metabolism , Animals , Brain/pathology , Genotype
16.
Eur J Neurosci ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38887188

ABSTRACT

Mouse neuronal CAD 5 cell line effectively propagates various strains of prions. Previously, we have shown that it can also be differentiated into the cells morphologically resembling neurons. Here, we demonstrate that CAD 5 cells chronically infected with prions undergo differentiation under the same conditions. To make our model more realistic, we triggered the differentiation in the 3D culture created by gentle rocking of CAD 5 cell suspension. Spheroids formed within 1 week and were fully developed in less than 3 weeks of culture. The mature spheroids had a median size of ~300 µm and could be cultured for up to 12 weeks. Increased expression of differentiation markers GAP 43, tyrosine hydroxylase, ß-III-tubulin and SNAP 25 supported the differentiated status of the spheroid cells. The majority of them were found in the G0/G1 phase of the cell cycle, which is typical for differentiated cells. Moreover, half of the PrPC on the cell membrane was N-terminally truncated, similarly as in differentiated CAD 5 adherent cells. Finally, we demonstrated that spheroids could be created from prion-infected CAD 5 cells. The presence of prions was verified by immunohistochemistry, western blot and seed amplification assay. We also confirmed that the spheroids can be infected with the prions de novo. Our 3D culture model of differentiated CAD 5 cells is low cost, easy to produce and cultivable for weeks. We foresee its possible use in the testing of anti-prion compounds and future studies of prion formation dynamics.

17.
BMC Biotechnol ; 24(1): 32, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750469

ABSTRACT

ß-TCP ceramics are versatile bone substitute materials and show many interactions with cells of the monocyte-macrophage-lineage. The possibility of monocytes entering microporous ß-TCP ceramics has however not yet been researched. In this study, we used a model approach to investigate whether monocytes might enter ß-TCP, providing a possible explanation for the origin of CD68-positive osteoclast-like giant cells found in earlier works.We used flow chambers to unidirectionally load BC, PRP, or PPP into slice models of either 2 mm or 6 mm ß-TCP. Immunofluorescence for CD68 and live/dead staining was performed after the loading process.Our results show that monocytes were present in a relevant number of PRP and BC slices representing the inside of our 2 mm slice model and also present on the actual inside of our 6 mm model. For PPP, monocytes were not found beyond the surface in either model.Our results indicate the possibility of a new and so far neglected constituent in ß-TCP degradation, perhaps causing the process of ceramic degradation also starting from inside the ceramics as opposed to the current understanding. We also demonstrated flow chambers as a possible new in vitro model for interactions between blood and ß-TCP.


Subject(s)
Calcium Phosphates , Ceramics , Monocytes , Monocytes/cytology , Ceramics/chemistry , Calcium Phosphates/chemistry , Humans , Bone Substitutes/chemistry , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Porosity
18.
RNA ; 28(4): 583-595, 2022 04.
Article in English | MEDLINE | ID: mdl-35046126

ABSTRACT

A critical step of pre-mRNA splicing is the recruitment of U2 snRNP to the branch point sequence of an intron. U2 snRNP conformation changes extensively during branch helix formation, and several RNA-dependent ATPases are implicated in the process. However, the molecular mechanisms involved remain to be fully dissected. We took advantage of the differential nucleotide triphosphates requirements for DExD/H-box enzymes to probe their contributions to in vitro spliceosome assembly. Both ATP and GTP hydrolysis support the formation of A-complex, indicating the activity of a DEAH-enzyme because DEAD-enzymes are selective for ATP. We immunodepleted DHX15 to assess its involvement, and although splicing efficiency decreases with reduced DHX15, A-complex accumulation incongruently increases. DHX15 depletion also results in the persistence of the atypical ATP-independent interaction between U2 snRNP and a minimal substrate that is otherwise destabilized in the presence of either ATP or GTP. These results lead us to hypothesize that DHX15 plays a quality control function in U2 snRNP's engagement with an intron. In efforts to identify the RNA target of DHX15, we determined that an extended polypyrimidine tract is not necessary for disruption of the atypical interaction between U2 snRNP and the minimal substrate. We also examined U2 snRNA by RNase H digestion and identified nucleotides in the branch binding region that become accessible with both ATP and GTP hydrolysis, again implicating a DEAH-enzyme. Together, our results demonstrate that multiple ATP-dependent rearrangements are likely involved in U2 snRNP addition to the spliceosome and that DHX15 may have an expanded role in maintaining splicing fidelity.


Subject(s)
Ribonucleoprotein, U2 Small Nuclear , Spliceosomes , Introns/genetics , RNA Precursors/metabolism , RNA Splicing , RNA, Small Nuclear/genetics , Ribonuclease H/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Spliceosomes/metabolism
19.
Histochem Cell Biol ; 161(5): 381-389, 2024 May.
Article in English | MEDLINE | ID: mdl-38282055

ABSTRACT

Breast cancer treatment encompasses various therapeutic modalities, including surgery, radiotherapy, and chemotherapy. Breast-conserving surgery has been an integral part of breast cancer management. However, radiotherapy, an important component of breast cancer management, can lead to complications, particularly fibrosis, affecting reconstructive surgery outcomes. We conducted an in vivo study using 48 female Wistar Albino rats, employing segmental mastectomy and radiotherapy to simulate post-mastectomy conditions. The rats were divided into six groups: control, mastectomy, mastectomy + radiotherapy, mastectomy + platelet-rich plasma (PRP) + radiotherapy, mastectomy + infliximab + radiotherapy, and mastectomy + infliximab + PRP + radiotherapy. Edema, hyperemia, inflammation, and fibrosis were assessed as indicators of tissue response. Histopathological analysis revealed that mastectomy + infliximab and mastectomy + infliximab + PRP groups showed significant reductions in fibrosis compared to other groups. Edema, hyperemia, and inflammation were also less severe in these groups compared to the control group. Radiotherapy-induced fibrosis is a major concern in breast reconstruction. Our study suggests that local PRP application and systemic infliximab administration, either alone or in combination, could mitigate the adverse effects of radiotherapy. This approach has the potential to improve reconstructive outcomes in patients undergoing or having the possibility to undergo radiotherapy. This is the first study showing the effectiveness of infliximab and PRP combination on wound healing. The provided experimental rat model might offer guidance for further research. This study provides insights into optimizing outcomes in reconstructive breast surgery, paving the way for further research and clinical studies.


Subject(s)
Breast Neoplasms , Fibrosis , Infliximab , Platelet-Rich Plasma , Rats, Wistar , Infliximab/therapeutic use , Animals , Platelet-Rich Plasma/metabolism , Female , Rats , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Breast Neoplasms/drug therapy , Mastectomy
20.
Neuropathol Appl Neurobiol ; 50(1): e12963, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38353056

ABSTRACT

AIM: CH1641 was discovered in 1970 as a scrapie isolate that was unlike all other classical strains of scrapie isolated so far. We performed bio-assays of CH1641 in mice in order to further characterise this specific isolate. METHODS: We inoculated the original CH1641 isolate into ovine and bovine prion protein (PrP) transgenic mice as well as wild-type mice. In addition, we performed cross- and back passages between the various mouse lines to examine if one identical prion strain was isolated in all mouse lines or whether multiple prion strains exist in CH1641. RESULTS: We report the first successful transmission of CH1641 to wild-type RIII mice and via RIII mice to wild-type VM mice. Unexpectedly, analysis of the protease-resistant prion protein (PrPres ) in wild-type mice showed a classical scrapie banding pattern differing from the banding pattern of the original CH1641 isolate. Cross- and back passages of CH1641 between the various mouse lines confirmed that the same prion strain had been isolated in all mouse lines. CONCLUSIONS: The CH1641 isolate consists of a single prion strain but its molecular banding pattern of PrPres differs between wild-type mice and PrP transgenic mice. Consequently, molecular banding patterns of PrPres should be used with caution in strain typing since they do not solely depend on the properties of the prion strain but also on the host prion protein.


Subject(s)
Prions , Scrapie , Mice , Animals , Cattle , Sheep , Prions/metabolism , Scrapie/metabolism , Prion Proteins/genetics , PrPSc Proteins/metabolism , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL