Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PNAS Nexus ; 3(1): pgad479, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38274120

ABSTRACT

Minor intron-containing genes (MIGs) account for <2% of all human protein-coding genes and are uniquely dependent on the minor spliceosome for proper excision. Despite their low numbers, we surprisingly found a significant enrichment of MIG-encoded proteins (MIG-Ps) in protein-protein interactomes and host factors of positive-sense RNA viruses, including SARS-CoV-1, SARS-CoV-2, MERS coronavirus, and Zika virus. Similarly, we observed a significant enrichment of MIG-Ps in the interactomes and sets of host factors of negative-sense RNA viruses such as Ebola virus, influenza A virus, and the retrovirus HIV-1. We also found an enrichment of MIG-Ps in double-stranded DNA viruses such as Epstein-Barr virus, human papillomavirus, and herpes simplex viruses. In general, MIG-Ps were highly connected and placed in central positions in a network of human-host protein interactions. Moreover, MIG-Ps that interact with viral proteins were enriched with essential genes. We also provide evidence that viral proteins interact with ancestral MIGs that date back to unicellular organisms and are mainly involved in basic cellular functions such as cell cycle, cell division, and signal transduction. Our results suggest that MIG-Ps form a stable, evolutionarily conserved backbone that viruses putatively tap to invade and propagate in human host cells.

2.
Mol Cell ; 83(12): 1983-2002.e11, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37295433

ABSTRACT

The evolutionarily conserved minor spliceosome (MiS) is required for protein expression of ∼714 minor intron-containing genes (MIGs) crucial for cell-cycle regulation, DNA repair, and MAP-kinase signaling. We explored the role of MIGs and MiS in cancer, taking prostate cancer (PCa) as an exemplar. Both androgen receptor signaling and elevated levels of U6atac, a MiS small nuclear RNA, regulate MiS activity, which is highest in advanced metastatic PCa. siU6atac-mediated MiS inhibition in PCa in vitro model systems resulted in aberrant minor intron splicing leading to cell-cycle G1 arrest. Small interfering RNA knocking down U6atac was ∼50% more efficient in lowering tumor burden in models of advanced therapy-resistant PCa compared with standard antiandrogen therapy. In lethal PCa, siU6atac disrupted the splicing of a crucial lineage dependency factor, the RE1-silencing factor (REST). Taken together, we have nominated MiS as a vulnerability for lethal PCa and potentially other cancers.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Introns/genetics , Prostatic Neoplasms/metabolism , RNA Splicing/genetics , Spliceosomes/metabolism , Signal Transduction , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Cell Line, Tumor , Prostatic Neoplasms, Castration-Resistant/genetics
3.
Development ; 148(20)2021 10 15.
Article in English | MEDLINE | ID: mdl-34557915

ABSTRACT

Minor spliceosome inhibition due to mutations in RNU4ATAC are linked to primary microcephaly. Ablation of Rnu11, which encodes a minor spliceosome snRNA, inhibits the minor spliceosome in the developing mouse pallium, causing microcephaly. There, cell cycle defects and p53-mediated apoptosis in response to DNA damage resulted in loss of radial glial cells (RGCs), underpinning microcephaly. Here, we ablated Trp53 to block cell death in Rnu11 cKO mice. We report that Trp53 ablation failed to prevent microcephaly in these double knockout (dKO) mice. We show that the transcriptome of the dKO pallium was more similar to the control compared with the Rnu11 cKO. We find aberrant minor intron splicing in minor intron-containing genes involved in cell cycle regulation, resulting in more severely impaired mitotic progression and cell cycle lengthening of RGCs in the dKO that was detected earlier than in the Rnu11 cKO. Furthermore, we discover a potential role of p53 in causing DNA damage in the developing pallium, as detection of γH2aX+ was delayed in the dKO. Thus, we postulate that microcephaly in minor spliceosome-related diseases is primarily caused by cell cycle defects.


Subject(s)
Introns/genetics , Microcephaly/genetics , RNA Splicing/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Cycle/genetics , Cell Death/genetics , Ependymoglial Cells/pathology , Female , Male , Mice , Mice, Knockout , Mutation/genetics , RNA, Small Nuclear/genetics , Spliceosomes/genetics , Transcriptome/genetics
4.
Development ; 147(21)2020 08 14.
Article in English | MEDLINE | ID: mdl-32665241

ABSTRACT

Disruption of the minor spliceosome due to mutations in RNU4ATAC is linked to primordial dwarfism in microcephalic osteodysplastic primordial dwarfism type 1, Roifman syndrome, and Lowry-Wood syndrome. Similarly, primordial dwarfism in domesticated animals is linked to positive selection in minor spliceosome components. Despite being vital for limb development and size regulation, its role remains unexplored. Here, we disrupt minor spliceosome function in the developing mouse limb by ablating one of its essential components, U11 small nuclear RNA, which resulted in micromelia. Notably, earlier loss of U11 corresponded to increased severity. We find that limb size is reduced owing to elevated minor intron retention in minor intron-containing genes that regulate cell cycle. As a result, limb progenitor cells experience delayed prometaphase-to-metaphase transition and prolonged S-phase. Moreover, we observed death of rapidly dividing, distally located progenitors. Despite cell cycle defects and cell death, the spatial expression of key limb patterning genes was maintained. Overall, we show that the minor spliceosome is required for limb development via size control potentially shared in disease and domestication.


Subject(s)
Dwarfism/genetics , Extremities/embryology , Fetal Growth Retardation/genetics , Microcephaly/genetics , Osteochondrodysplasias/genetics , RNA, Small Nuclear/metabolism , Animals , Body Patterning/genetics , Cell Cycle/genetics , Female , Forelimb/embryology , Forelimb/ultrastructure , Gene Expression Regulation, Developmental , Hindlimb/embryology , Hindlimb/ultrastructure , Introns/genetics , Male , Mice, Inbred C57BL , Mutation/genetics , RNA, Small Nuclear/genetics , Stem Cells/metabolism
5.
Front Genet ; 10: 1113, 2019.
Article in English | MEDLINE | ID: mdl-31798628

ABSTRACT

Minor introns constitute <0.5% of the introns in the human genome and have remained an enigma since their discovery. These introns are removed by a distinct splicing complex, the minor spliceosome. Both are ancient, tracing back to the last eukaryotic common ancestor (LECA), which is reflected by minor intron enrichment in specific gene families, such as the mitogen activated-protein kinase kinases, voltage-gated sodium and calcium ion channels, and E2F transcription factors. Most minor introns occur as single introns in genes with predominantly major introns. Due to this organization, minor intron-containing gene (MIG) expression requires the coordinated action of two spliceosomes, which increases the probability of missplicing. Thus, one would expect loss of minor introns via purifying selection. This has resulted in complete minor intron loss in at least nine eukaryotic lineages. However, minor introns are highly conserved in land plants and metazoans, where their importance is underscored by embryonic lethality when the minor spliceosome is inactivated. Conditional inactivation of the minor spliceosome has shown that rapidly dividing progenitor cells are highly sensitive to minor spliceosome loss. Indeed, we found that MIGs were significantly enriched in a screen for genes essential for survival in 341 cycling cell lines. Here, we propose that minor introns inserted randomly into genes in LECA or earlier and were subsequently conserved in genes crucial for cycling cell survival. We hypothesize that the essentiality of MIGs allowed minor introns to endure through the unicellularity of early eukaryotic evolution. Moreover, we identified 59 MIGs that emerged after LECA, and that many of these are essential for cycling cell survival, reinforcing our essentiality model for MIG conservation. This suggests that minor intron emergence is dynamic across eukaryotic evolution, and that minor introns should not be viewed as molecular fossils. We also posit that minor intron splicing was co-opted in multicellular evolution as a regulatory switch for en masse control of MIG expression and the biological processes they regulate. Specifically, this mode of regulation could control cell proliferation and thus body size, an idea supported by domestication syndrome, wherein MIGs are enriched in common candidate animal domestication genes.

6.
Development ; 145(17)2018 08 28.
Article in English | MEDLINE | ID: mdl-30093551

ABSTRACT

Mutation in minor spliceosome components is linked to the developmental disorder microcephalic osteodysplastic primordial dwarfism type 1 (MOPD1). Here, we inactivated the minor spliceosome in the developing mouse cortex (pallium) by ablating Rnu11, which encodes the crucial minor spliceosome small nuclear RNA (snRNA) U11. Rnu11 conditional knockout mice were born with microcephaly, which was caused by the death of self-amplifying radial glial cells (RGCs), while intermediate progenitor cells and neurons were produced. RNA sequencing suggested that this cell death was mediated by upregulation of p53 (Trp53 - Mouse Genome Informatics) and DNA damage, which were both observed specifically in U11-null RGCs. Moreover, U11 loss caused elevated minor intron retention in genes regulating the cell cycle, which was consistent with fewer RGCs in S-phase and cytokinesis, alongside prolonged metaphase in RGCs. In all, we found that self-amplifying RGCs are the cell type most sensitive to loss of minor splicing. Together, these findings provide a potential explanation of how disruption of minor splicing might cause microcephaly in MOPD1.


Subject(s)
Cell Cycle/genetics , Cell Death/physiology , Dwarfism/genetics , Ependymoglial Cells/metabolism , Fetal Growth Retardation/genetics , Microcephaly/genetics , Neural Stem Cells/cytology , Osteochondrodysplasias/genetics , RNA Splicing/genetics , RNA, Small Nuclear/genetics , Spliceosomes/genetics , Animals , Base Sequence , Mice , Mice, Inbred C57BL , Mice, Knockout , Spliceosomes/metabolism , Tumor Suppressor Protein p53/biosynthesis
7.
Appl Opt ; 56(33): 9277-9284, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-29216100

ABSTRACT

Microplasma is generated in an ultra-high-pure H2 and N2 gas mixture with a Nd:YAG laser device that is operated at the fundamental wavelength of 1064 nm. The gas mixture ratio of H2 and N2 is 9 to 1 at a pressure of 1.21 ± 0.03 105 Pa inside a chamber. A Czerny-Turner-type spectrometer and an intensified charge-coupled device are utilized for the recording of plasma emission spectra. The line-of-sight measurements are Abel inverted to determine the radial distributions of electron number density and temperature. Recently derived empirical formulas are utilized for the extraction of values for electron density. The Boltzmann plot and line-to-continuum methods are implemented for the diagnostic of electron excitation temperature. The expansion speed of the plasma kernel maximum electron temperature amounts to 1 km/s at a time delay of 300 ns. The microplasma, initiated by focusing 14 ns, 140 mJ pulses, can be described by an isentropic expansion model.

SELECTION OF CITATIONS
SEARCH DETAIL
...