Poster abstracts

Log in and return to this page to see the full titles and abstracts.

1. The h18pep1 inhibit RsuA binding to 16S rRNA

Sudeshi M. Abedeera (Department of Chemistry and Biochemistry, Kent State University), Jiale Xie (Department of Chemistry and Biochemistry, Kent State University), Sanjaya C. Abeysirigunawardena (Department of Chemistry and Biochemistry, Kent State University)

Abstract:
Ribosomes are an essential cellular component that carries out protein biosynthesis in all forms of life. Bacterial ribosome modification enzymes can have important biological roles other than their primary RNA modification function. Although many bacterial pseudouridine synthases are found to be non-essential for bacterial survival under normal growth conditions, their presence may be critical under non-conducing conditions such as environmental stress. Our studies illustrate that RsuA plays a crucial role in bacterial survival under streptomycin stress. Furthermore, the pseudouridylation activity of RsuA was observed to be important to render the survival advantage against streptomycin. We hypothesized that the efficacy of streptomycin as an antibiotic can be enhanced by blocking RsuA binding at helix18 thereby preventing the pseudouridylation at U516. We used phage display technique to discover a 12-mer peptide (h18pep1) that specifically binds to the same region of helix18 to which RsuA is known to interact. FRET-based assays were carried out using helix18 model RNA and 16S 5´-domain RNA to determine the ability of the selected 12-mer peptide to prevent RsuA binding to helix18. This work illustrates a useful strategy to combat antibiotic resistance in bacteria.

Keywords: Antibiotic resistance, Pseudouridine synthase, Streptomycin

2. Human trans-editing enzyme displays tRNA acceptor stem specificity and relaxed amino acid selectivity

Oscar Vargas-Rodriguez (Department of Chemistry and Biochemistry and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA.), Marina Bakhtina (Department of Chemistry and Biochemistry and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA.), Daniel McGowan (Department of Chemistry and Biochemistry and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA.), Jawad Abid (Department of Chemistry and Biochemistry and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA.), Hiroaki Suga (Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan.), Karin Musier-Forsyth (Department of Chemistry and Biochemistry and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA.)

Abstract:
Aminoacyl-tRNA synthetases (ARSs) are a group of ancient enzymes responsible for pairing amino acids (aa) with their cognate tRNAs. Due to similarity in sizes and chemical properties of aa, ARSs are prone to errors in aminoacylation. Prolyl-tRNA synthetase (ProRS) mischarges Ala on to tRNAPro, which can be detrimental to translation fidelity and cell viability. These errors can be corrected by an editing domain present in some ProRS or by free standing trans-editing proteins. We previously showed that the Caulobacter crescentus (Cc) ProXp-ala uses a size-based exclusion mechanism and conformational selection to specifically recognize Ala-tRNAPro. In the bacterial system, tRNA acceptor stem bases G72 and A73 act as recognition elements for ProXp-ala. However, the mechanism by which Homo sapiens (Hs) ProXp-ala recognizes aa-tRNA remained to be explored. Here, we report that Hs ProXp-ala is also an efficient Ala-tRNAPro deacylase and has strong recognition elements in the Hs tRNAPro acceptor stem including bases C72 and C73. Mutating conserved residues in the substrate binding pocket of Hs and Cc ProXp-ala modulates the aa selectivity of these enzymes. Our results reveal the evolutionary divergence between bacterial and Hs ProXp-ala in their tRNA and aa selectivity and sheds light on how these proteins coevolved with their tRNA substrates. Taken together, these results illustrate how the mechanism of substrate selection diverged during the evolution of the ProXp-ala family and provide the first example of a trans-editing domain whose specificity evolved to adapt to the changes in its tRNA substrate.

References:
Vargas-Rodriguez, O., Bakhtina, M., McGowan, D., Abid, J., Goto, Y., Suga, H., & Musier-Forsyth, K. (2020). Human trans-editing enzyme displays tRNA acceptor-stem specificity and relaxed amino acid selectivity. Journal of Biological Chemistry, 295(48), 16180-16190.

Keywords: Aminoacyl-tRNA synthetases, tRNA editing

3. Title not available online - please see the booklet.

Hannah R. Ackerman (Nationwide Childrens Center for Childhood Cancer and Ohio State University Pediatrics ), Dawn S Chandler (Nationwide Childrens Center for Childhood Cancer), Matias Montes (Nationwide Childrens Center for Childhood Cancer)

Abstract not available online - please check the booklet.

4. Unraveling the roles of 5’ transcript leaders in gene regulation

Christina Akirtava (Carnegie Mellon University, Biological Sciences), Hunter Kready (Carnegie Mellon University, Biological Sciences), Lauren Nazzaro (Carnegie Mellon University, Biological Sciences), Matt Agar-Johnson (Carnegie Mellon University, Biological Sciences), Gemma E May (Carnegie Mellon University, Biological Sciences), C. Joel McManus (Carnegie Mellon University, Biological Sciences)

Abstract not available online - please check the booklet.

5. TCAB1 is necessary for telomerase RNP assembly

Basma Al Masraf (Cellular and Molecular Biology Graduate Program, Michigan State University), Gloria I. Perez (Institute for Quantitative Health Sciences and Engineering, Michigan State University), Kate Adams-Boone (Institute for Quantitative Health Sciences and Engineering, Michigan State University), Scott B. Cohen (Childrens Medical Research Institute and University of Sydney), Li Han (Department of Microbiology and Molecular Genetics, Michigan State University), Kefei Yu (Department of Microbiology and Molecular Genetics, Michigan State University)

Abstract:
The ribonucleoprotein telomerase counteracts telomere shortening by adding repetitive sequences to the ends of human chromosomes. Telomerase is composed of the reverse transcriptase TERT, the telomerase RNA, and several auxiliary proteins that associate with the telomerase RNA, including TCAB1. TCAB1 is necessary for telomere maintenance in human cells and has been proposed to play a role in telomerase trafficking to Cajal bodies and telomeres, and in telomerase RNA folding. Here we show that, contrary to previous findings, TCAB1 is essential for telomerase assembly. We demonstrate that in the absence of TCAB1, the telomerase RNA is trapped in the nucleolus, a phase separated nuclear organelle, while TERT localizes to the nucleoplasm and is excluded from the nucleolus. Thus, nucleolar phase separation constitutes a barrier that counteracts telomerase assembly and TCAB1 is required to extract the telomerase RNA from the nucleolus, providing a molecular mechanism for the essential role of TCAB1 in telomerase function.

References:
Armanios, M., and Blackburn, E.H. (2012). The telomere syndromes. Nat Rev Genet 13, 693 704.
Chen, L., Roake, C.M., Freund, A., Batista, P.J., Tian, S., Yin, Y.A., Gajera, C.R., Lin, S., Lee, B., Pech, M.F., et al. (2018). An Activity Switch in Human Telomerase Based on RNA Conformation and Shaped by TCAB1. Cell 174.
Cohen, S.B., Graham, M.E., Lovrecz, G.O., Bache, N., Robinson, P.J., and Reddel, R.R. (2007). Protein Composition of Catalytically Active Human Telomerase from Immortal Cells. Science 315, 1850–1853.
Cong, L., Ran, F.A., Cox, D., Lin, S., Barretto, R., Habib, N., Hsu, P.D., Wu, X., Jiang, W., Marraffini, L.A., et al. (2013). Multiplex Genome Engineering Using CRISPR/Cas Systems. Science 339, 819–823.
Cristofari, G., and Lingner, J. (2006). Telomere length homeostasis requires that telomerase levels are limiting. Embo J 25, 565 574.

Keywords: Telomerase RNP, TCAB1, Telomerase RNA

6. Expanding native IM-MS into the transcriptome

Anna G. Anders (Department of Chemsitry), Elizabeth E. Tidwell (Department of Biophysics), Varun Gadkari (Department of Chemistry), Markos Koutmos (Department of Biophysics), Brandon T. Ruotolo (Department of Chemistry)

Abstract not available online - please check the booklet.

7. Investigating Regulators of Nonsense Mediated mRNA Decay in Zebrafish Neuromuscular Development

Rene Arvola (Department of Molecular Genetics, The Ohio State University), Zhongxia Yi, Thomas Gallagher (Department of Molecular Genetics, The Ohio State University), Amy Everest (Department of Neuroscience, The Ohio State University ), Pooja Gangras (Department of Molecular Genetics, The Ohio State University), Sharon Amacher (Department of Molecular Genetics, Department of Biological Chemistry and Pharmacology, The Ohio State University), Guramrit Singh (Department of Molecular Genetics, The Ohio State University)

Abstract not available online - please check the booklet.

8. Investigating the epitranscriptome of the early embryo

Taylor N. Ayers (University of Pittsburgh ), Joel C. Rosenbaum (University of Pittsburgh), Hannah E. Moore (University of Pittsburgh), Wesley A. Phelps (University of Pittsburgh), Anne E. Carlson (University of Pittsburgh), Miler T. Lee (University of Pittsburgh )

Abstract not available online - please check the booklet.

9. ESRP2–microRNA-122 axis directs the postnatal onset of liver maturation and polyploidization.

Sushant Bangru (University of Illinois Urbana-Champaign, Department of Biochemistry, Urbana, IL)

Abstract not available online - please check the booklet.

10. Functional characterization and therapeutic opportunities of RNA splicing factor mutants in myelodysplastic syndromes

Maxwell Bannister (Department of Pharmacology, University of Minnesota), Chan Zhou (Department of Population and Quantitative Health Sciences, University of Massachusetts ), Stanley Lee (Clinical Research Division, Fred Hutchinson Cancer Research Center), Hai Dang Nguyen (Department of Pharmacology, University of Minnesota)

Abstract:
Somatic heterozygous mutations in the RNA splicing factor (SF) genes U2AF1, SF3B1, SRSF2, and ZRSR2 occur in >50% of patients with myelodysplastic syndromes (MDS), a hematological malignancy that often progresses to secondary acute myeloid leukemia (sAML). Mutant SFs cause transcriptome-wide mis-splicing of pre-mRNA, but how this contributes to MDS pathogenesis is unclear. Moreover, targeting pre-mRNA splicing in patients carrying a SF mutation has been ineffective. My research aimed to identify new mechanistic rationales and potential targets for therapeutic development in MDS patients harboring a SF mutation. First, we found that the U2AF1S34F mutant induces the differential expression of circular RNAs (circRNAs), an emerging class of non-coding RNA formed by the ‘backsplicing’ of exons from a single pre-mRNA. Future work will characterize the disease relevance of these circRNAs. Second, our lab reported that SF-mutant cells accumulate aberrant levels of R-loops, three-stranded nucleic acid structures containing an RNA:DNA hybrid and a displaced single-stranded DNA. We hypothesized that inhibiting R-loop resolution would selectively kill SF-mutant cells. To identify potential targets, a fluorogenic high-throughput screening (HTS) assay was developed to measure RNA:DNA hybrid resolution in real-time. In parallel, we developed an in-house computational pipeline for drug sensitivity analysis and screened a small panel of compounds.

Keywords: RNA splicing, circular RNA, Myelodysplastic syndromes

11. Rational Design for Controlled Release of Dicer Substrate siRNA Harbored in phi29 Based RNA Nanoparticles

Daniel W Binzel (Center for RNA Nanobiotechnology and Nanomedicine; Division of Pharmaceutics and Pharmacology, College of Pharmacy; Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University), Songchuan Guo (Center for RNA Nanobiotechnology and Nanomedicine; Division of Pharmaceutics and Pharmacology, College of Pharmacy; Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University), Hongran Yin (Center for RNA Nanobiotechnology and Nanomedicine; Division of Pharmaceutics and Pharmacology, College of Pharmacy; Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University), Tae Jin Lee (Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston), Shujun Liu (The Hormel Institute, Masonic Cancer Center, University of Minnesota), Peixuan Guo (Center for RNA Nanobiotechnology and Nanomedicine; Division of Pharmaceutics and Pharmacology, College of Pharmacy; Dorothy M. Davis Heart and Lung Research Institute; The Ohio State University)

Abstract not available online - please check the booklet.

12. Using SMN mutants to determine what functions SMN performs and which are critical to SMA

Anton J. Blatnik III (Department of Biological Chemistry & Pharmacology, The Ohio State University Wexner Medical Center), Vicki L. McGovern, Corey Ruhno, Arthur H.M. Burghes (Department of Biological Chemistry & Pharmacology, The Ohio State University Wexner Medical Center), Veronica Pessino, Shawn Driscoll, Samuel Pfaff (Gene Expression Laboratory, The Salk Institute for Biological Studies), Utz Fischer (Department of Biochemistry, The University of Wurzburg), Shibi B. Likhite, Kathrin C. Meyer (Department of Gene Therapy, The Abigail Wexner Research Institute at Nationwide Childrens Hospital)

Abstract:
How survival motor neuron (SMN) protein deficiency results in motor neuron loss in spinal muscular atrophy (SMA) is currently unknown. SMN is an essential protein, whose canonical function is assembling the Sm-ring onto the spliceosomal small nuclear ribonucleic acids (snRNAs), but other functions have been proposed.
We have developed a cell assay that can conditionally delete wild-type SMN production using Cre-recombinase, resulting in cell death. Using these cells, we have determined that all disease causing SMN missense mutations in are incapable of essential function on their own. Additionally, an SMN lacking exon 2B is capable of essential function and snRNP assembly. Residues in exon 2B are reported to bind the COPI subunit alpha-COP, important for transmembrane protein shuttling and a suggested mechanism of RNP trafficking down motor neuron axons. SMA mice injected with an AAV9 expressing the SMN lacking exon 2B show rescued motor units and increased survival, suggesting the alpha-COP association is not critical to SMA.
Also, we have performed a suppressor screen to determine the lost function of the SMNE134K mutation and confirmed a mutant Sm protein as a suppressor. This implicates Sm-assembly as the essential function of SMN and in vitro snRNP assembly assays indicate, together these mutants perform assembly more efficiently than wild-type conditions. We have created SMA mice that transgenically express SMNE134K and will treat them with an AAV9 expressing the Sm-suppressor to determine if Sm-assembly is the critical function of SMN pertaining to SMA. We are also investigating whether the SMNE134K and Sm suppressor combinantion retains fidelity of snRNP assembly.
These experiments will determine what functions SMN performs and whether they play a critical role in the development of SMA. This may reveal a mechanism by which mutations in a ubiquitous RNP assembler gives rise to a cell specific disease phenotype.

References:
Blatnik, A.J., McGovern, V.L., Le, T.T., Iyer, C.C., Kaspar, B.K. and Burghes, A.H.M. (2020) Conditional deletion of SMN in cell culture identifies functional SMN alleles. Hum Mol Genet, 29, 3477-3492.
Custer, S.K., Todd, A.G., Singh, N.N. and Androphy, E.J. (2013) Dilysine motifs in exon 2b of SMN protein mediate binding to the COPI vesicle protein alpha-COP and neurite outgrowth in a cell culture model of spinal muscular atrophy. Hum Mol Genet, 22, 4043-4052.
Li, H., Custer, S.K., Gilson, T., Hao le, T., Beattie, C.E. and Androphy, E.J. (2015) alpha-COP binding to the survival motor neuron protein SMN is required for neuronal process outgrowth. Hum Mol Genet, 24, 7295-7307.

Keywords: SMN, Sm-assembly, suppressor screen

13. Drug Discovery Studies Targeting the Stem Loop 2 Motif in SARS-CoV-2 RNA

H. Boesger (Department of Biological Sciences, Honors Tutorial College, Ohio University ), Md. I. Hossain (Department of Chemistry & Biochemistry, Ohio University ), E. Marino (Department of Biological Sciences, Honors Tutorial College, Ohio University), M. Myers (Department of Chemistry & Biochemistry, Honors Tutorial College, Ohio University ), E. Fairchild, A.H. Aldhumani (Department of Chemistry & Biochemistry, Ohio University ), J.V. Hines (Department of Chemistry & Biochemistry, Ohio University )

Abstract not available online - please check the booklet.

14. Identification of a tRNA-specific function for the tRNA methyltransferase Trm10 in Saccharomyces cerevisiae

Isobel Bowles (OSBP), Jane Jackman (Ohio State Chemistry and Biochemistry )

Abstract:
tRNA methyltransferase 10 (Trm10) methylates N1 of guanosine at the 9th position of tRNA molecules using methyl donor S-adenosyl methionine (SAM). Upon deletion of TRM10, Saccharomyces cerevisiae strains exhibit growth defects in the presence of antitumor drug 5-fluorouracil (5FU). We hypothesized that tRNA stability decreases with the lack of the m1G9 modification in trm10Δ strains and that certain tRNA species are more reliant upon the presence of the methylated G9 nucleotide, as evident from different growth phenotypes observed upon tRNA overexpression. When Trm10 substrate tRNATrp is overexpressed in trm10Δ strains, growth hypersensitivity to 5FU is rescued, while overexpression of other tRNA species in S. cerevisiae do not display a growth rescue. We demonstrated that levels of tRNATrp decrease in trm10Δ strains and that overexpression of tRNATrp recovers tRNATrp levels in trm10Δ strains, while another Trm10 substrate (tRNAGly) remains at a similar level in all strains. The specific role of the m1G9 tRNA modification is being investigated by analyzing known mechanisms of tRNATrp quality control, using S. cerevisiae trm10Δ strains with 5FU. tRNA structures that correlate with active substrates for Trm10 activity are also being determined with nuclease and chemical footprinting, as well as 2’ hydroxyl acylation analyzed by primer extension (SHAPE). Together these studies provide further understanding of tRNA structural elements that promote methylation by Trm10 and the biological impact of loss of this highly conserved modification.

Keywords: tRNA modification, tRNA levels, SHAPE

15. Expression platform features tune functional characteristics of the Clostridium beijerincki ZTP riboswitch

David Z. Bushhouse (Interdisciplinary Biological Sciences Graduate Program, Northwestern University; Center for Synthetic Biology, Northwestern University), Julius B. Lucks (Center for Synthetic Biology, Northwestern University; Department of Chemical and Biological Engineering, Northwestern University)

Abstract:
Riboswitches are short cis-encoded RNA elements that regulate the expression of downstream genes in response to small molecule ligands.1 While much is known about the structural nature of riboswitch-ligand interactions, we are only beginning to understand how the dynamic process of co-transcriptional RNA folding leads to regulatory outcomes.2 To elucidate principles of expression platform function, we are investigating the Clostridium beijerincki ZTP riboswitch, a transcriptional ON switch that senses ZTP (~115μM EC50) as an indicator of folate stress.3 In the absence of ZTP/ZMP, the formation of a strong intrinsic terminator attenuates transcription; however, in the presence of ZTP/ZMP, the formation of this hairpin is delayed by a ligand-dependent alternative folding pathway, allowing RNA polymerase to process past the termination site.4 Previous co-transcriptional structure probing studies implicated strand displacement as the primary gene regulatory mechanism. How the strand displacement conformational change is controlled by the expression platform, however, remains poorly understood.4 Using an in vivo reporter assay and a functional mutagenesis approach, we have found that the length of the terminator hairpin loop can widely tune riboswitch sensitivity (~30–220 μM EC50), extensions to the terminator hairpin affect dynamic range (~3–15-fold), and strategically placed mismatches and bulges within the terminator stem can widely alter baseline expression and dynamic range (~2–19-fold). Together, our findings show that the Cbe ZTP expression platform tunes the functional characteristics of the riboswitch via a concerted combination of sequence and structural elements to achieve the baseline expression, sensitivity, and maximal response amenable to the organism. These results also indicate possible approaches for the optimization of natural riboswitches that employ strand displacement mechanisms for biotechnology applications.

References:
1. Breaker, R. R. Riboswitches and the RNA world. Cold Spring Harbor Perspectives in Biology 4, 1–15 (2012).
2. Watters, K. E., Strobel, E. J., Yu, A. M., Lis, J. T. & Lucks, J. B. Cotranscriptional folding of a riboswitch at nucleotide resolution. Nature Structural and Molecular Biology 23, 1124–1131 (2016).
3. Kim, P. B., Nelson, J. W. & Breaker, R. R. An ancient riboswitch class in bacteria regulates purine biosynthesis and one-carbon metabolism. Molecular Cell 57, 317–328 (2015).
4. Strobel, E. J., Cheng, L., Berman, K. E., Carlson, P. D. & Lucks, J. B. A ligand-gated strand displacement mechanism for ZTP riboswitch transcription control. Nature Chemical Biology 15, 1067–1076 (2019).

Keywords:

16. Synergistic properties of ribosome-targeting macrolides and hygromycin A

Chih Wei Chen (University of Illinois at Chicago), Yury Polikanov (University of Illinois at Chicago)

Abstract:
Antibiotic resistance is one of the fastest-growing and significant issue in healthcare. Bacteria can employ various methods to circumvent the effects of antibiotics that bind to the ribosome, the main target for antibacterial drugs. One approach to overcome antibiotic resistance is utilizing synergistic combinations of antibiotics. Currently, there are limited pairings of ribosome-targeting antibiotics that exhibit synergy. In this work, we discovered a novel combination of antibiotics (hygromycin A/macrolide) that display cooperativity in ribosome binding as well as antimicrobial function. We obtained crystal structures of hygromycin A bound simultaneously with one of three different macrolides (Erythromycin, azithromycin and telithromycin) in complex with Thermus thermophilus 70S ribosome at a high resolution (2.45 to 2.6Å). Our structures allows us to elucidate the mechanism of action of the observed binding and antimicrobial cooperativity. Through our study, we provide a new direction for the development and discovery of synergistic antibiotic tandems.

Keywords: Ribosome, Macrolides, Hygromycin A

18. Dynamic expression of poly(A) binding protein N1 coordinates cardiac growth during development and disease

Joe Seimetz,Sandip Chorghade, Bo Zhang, Chaitali Misra (Department of Biochemistry , University of Illinois Urbana-Champaign), Stefan Bresson,Nicholas Conrad (Department of Microbiology, University of Texas Southwestern Medical Center), Xander Wehrens (Department of Molecular Physiology and Biophysics, Baylor College of Medicine), Auinash Kalsotra (Department of Biochemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign)

Abstract:
Global protein synthesis rates vary among tissues and cell types depending on their function and environment, and this variation is essential for normal development, growth, and homeostasis. Although the protein synthesis rate in the adult heart is one of the lowest among all tissues, it increases drastically in response to hypertrophic stimuli. Post-transcriptional changes can greatly affect translation of mRNAs. Particularly, the 3’UTR sequence and poly(A) tail length can influence translation through RNA binding proteins and non-coding RNAs. Nuclear poly(A) binding protein (PABPN1) is one such RBP that regulates mRNA translation via poly(A) tail length, mRNA transport, and alternative polyadenylation (ApA).
Here we report that cardiac PABPN1 protein levels are drastically downregulated during postnatal heart development. Strikingly, this down-regulation is post-transcriptional, cardiomyocyte-specific, evolutionarily conserved, and reversed with heart disease. Reduced PABPN1 protein levels in adult cardiomyocytes correlate strongly with the decrease in total protein synthesis, widespread changes in ApA, and low abundance of transcripts with longer poly(A) tails. Furthermore, we discovered that PABPN1-depleted cardiomyocytes are viable; but refractory to stimulus-induced hypertrophic growth due to their inability to activate new protein synthesis. Remarkably, forced expression of PABPN1 in the adult myocardium of transgenic mice stimulates pathological cardiac hypertrophy by modulating ApA, poly(A) tail length, and translation of mRNAs that are directly linked to maladaptive cardiac growth and heart failure. These data demonstrate that dynamic regulation PABPN1 facilitates transcriptome remodeling, protein synthesis, and subsequent growth of the mammalian heart in response to developmental and hypertrophic signals.

Keywords: PABPN1, Polyadenylation, Hypertrophy

19. tRNA family-specific turnover pathways and environmental stresses regulate tRNA intron degradation

Katherine Clark (Department of Molecular Genetics, Center for RNA Biology, OSU, Columbus, OH), Sara Metcalf (Department of Molecular Genetics, Center for RNA Biology, OSU, Columbus, OH), Alicia Bao (Department of Molecular Genetics, Center for RNA Biology, OSU, Columbus, OH), Lauren Peltier (Department of Molecular Genetics, Center for RNA Biology, OSU, Columbus, OH), Anita K. Hopper (Department of Molecular Genetics, Center for RNA Biology, OSU, Columbus, OH)

Abstract:
In S. cerevisiae, 10 tRNA families are encoded by intron-containing genes. Precursor tRNAs are processed into mature functional tRNAs via several steps, including intron removal by the tRNA splicing endonuclease complex. After splicing, the free introns are rapidly and efficiently degraded. The mechanism for tRNA intron turnover has been delineated for two of the 10 families; tRNAIleUAU and tRNALeuCAA linear introns are degraded by the 5’ to 3’ exonuclease Xrn1 after 5’ phosphorylation by the tRNA ligase/kinase Rlg1(Trl1). There are at least 4 additional pathways for the remaining 8 tRNA families: Xrn1-independent, Rlg1-independent, Xrn1 and Rlg1-independent, and an endonuclease-dependent pathway for the circular form of the tRNATrp intron. To discover gene products functioning in these unknown pathways, we conducted a candidate screen of 60 mutant S. cerevisiae strains that are defective in annotated genes involved in RNA degradation and/or RNA phosphorylation. We identified several candidates as participants in tRNA family-specific intron turnover, including the 3’ to 5’ nuclease Rrp4, the endonuclease Las1, and the 5’ to 3’ nuclease regulator Met22. We also investigated the effects of various environmental stresses upon tRNA intron levels and surprisingly uncovered stress-specific, tRNA family-specific intron accumulation. Treatment of wild-type cells with 3mM H2O2 causes 15- and 8-fold elevations of tRNATrpCCA and tRNALeuCAA intron levels, respectively. Accumulation of these tRNA introns occurs for >2 hours, despite immediate termination of tRNA transcription upon oxidative stress. Inversely, most tRNA intron levels decrease with 5% ethanol or 42˚C heat treatment. One exception is tRNAProUGG which accumulates an aberrantly migrating intron upon heat stress. Our studies provide insight into family-specific endogenous and stress-induced intron turnover pathways that indicate possible novel regulatory functions of tRNA introns.

References:
1. Wu, J., & Hopper, A. K. (2014). Healing for destruction: tRNA intron degradation in yeast is a two-step cytoplasmic process catalyzed by tRNA ligase Rlg1 and 5'-to-3' exonuclease Xrn1. Genes & development, 28(14), 1556–1561. https://doi.org/10.1101/gad.244673.114

Keywords: tRNA turnover, intron degradation, environmental stress

20. A phosphorylation circuitry of RNaseH1 regulates R-loop homeostasis

Victor Corral (Pharmacology, University of Minnesota - Twin Cities), Hai D. Nguyen (Pharmacology, University of Minnesota - Twin Cities), Maxwell Bannister (Undergraduate studies, Augsburg University)

Abstract:
R-loop is a transcription intermediate resulting from the formation of stable RNA:DNA hybrids and a displaced single-stranded DNA (ssDNA). Aberrant accumulation or distribution of R-loops in the genome, if not regulated, becomes a source of genomic instability, a hallmark in many cancers. Our lab previously found that the ssDNA-binding protein, Replication Protein A (RPA), is a sensor of R-loops. RPA recruits and stimulates the activity of RNaseH1, an enzyme that specifically hydrolyzes the RNA moiety within RNA:DNA hybrids. How RPA-RNaseH1 interaction is regulated in cells is not known. Here, we provide evidence that phosphorylation of RNaseH1 is crucial for RPA-RNaseH1 interaction. RNaseH1 phospho-mutant at serine 76 (S76A), but not the phospho-mimetic mutant (S76E), disrupts its interaction with RPA. Importantly, expression of RNaseH1-S76A mutant, but not RNaseH1-S76E mutant, failed to suppress R-loop-associated genomic instability. Future studies will investigate how RNaseH1 phosphorylation affects its enzymatic activity and cellular functions. Altogether, these findings highlight a new phosphorylation circuitry by an unknown kinase critical for R-loop resolution through RNaseH1 function.

Keywords: RNaseH1, R-loop, Phosphorylation

21. Insights into RNA-protein interactions in Trypanosoma brucei telomerase

Justin A. Davis (University of North Carolina at Charlotte ), Kausik Chakrabarti (University of North Carolina at Charlotte )

Abstract:
Telomeres are the nucleoprotein structures found at the end of eukaryotic linear chromosomes. Because conventional DNA polymerases are unable to fully replicate the ends of linear chromosomes, the activity of the ribonucleoprotein enzyme telomerase is required to counteract the progressive loss of DNA after every cell division. The telomerase enzyme consists of two main components: the telomerase reverse transcriptase (TERT) protein and the telomerase RNA (TR), which provides the template for telomeric DNA synthesis. The TR forms a large structural scaffold on which accessory proteins can bind in order to form the complete telomerase holoenzyme in vivo. These accessory proteins are required for proper telomerase activity and regulation inside of cells. Interacting partners of TERT have been extensively characterized in yeast, human, and Tetrahymena systems. These interactors have not been extensively studied in lower eukaryotes including clinically relevant human parasites like Trypanosoma brucei (T. brucei), a causative agent of African sleeping sickness in humans. These parasites require constant telomerase activity in order to rapidly divide in their host and establish a chronic infection. In this study, we determined the telomerase interactome of the bloodstream forms of T. brucei by immunoaffinity purification and mass spectrometry. In addition, to investigate the effect of these interactions in the main metabolic pathways, we have determined the transcriptional response of these interactors to telomerase deficiency by real-time quantitative gene expression analysis. This data will provide us novel insights into telomerase regulation in these parasitic protozoa.

Keywords: RNA-protein interactions , Telomerase , Interactome

22. Role of the long non-coding RNA PACER in regulation of COX-2 gene expression

Joseph R. Iacona (Microbiology, Biochemistry and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School and the School of Graduate Studies ), Samuel Z. Desind (Microbiology, Biochemistry and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School and the School of Graduate Studies ), Carol S. Lutz (Microbiology, Biochemistry and Molecular Genetics, Rutgers Biomedical and Health Sciences, New Jersey Medical School and the School of Graduate Studies )

Abstract not available online - please check the booklet.

23. Myotonic Dystrophy Type 1 Adversely Alters the Adult Hepatocellular Transcriptome

Zachary Dewald (Biochemistry-University of Illinois Urbana Champaign), Auinash Kalsotra (Biochemistry-University of Illinois Urbana Champaign)

Abstract:
Myotonic Dystrophy type 1 (DM1) is multi-systemic muscular dystrophy, affecting 1 in 3000 people. DM1 is caused by a (CTG)n repeat expansion in the 3’ UTR of the ubiquitously expressed gene DMPK. The (CUG)n containing RNAs resulting from the transcription of this diseased DMPK gene aggregate in the nucleus, forming foci which sequester various RNA binding proteins (RBPs). The most pivotal RBPs sequestered are the muscleblind-like (MBNL) family proteins, a group of splicing factors that play significant roles in the juvenile-to-adult development of many tissues. Their sequestration in DM1 severely inhibits this maturation process in many tissues. Recent studies show that DM1 patients have increased susceptibility toward glucose intolerance, non-alcoholic fatty liver disease (NAFLD), and metabolic syndrome. Furthermore, DM1 patients are abnormally sensitive to various analgesics and anesthetics, with complications ranging from prolonged anesthesia recovery to heightened pulmonary dysfunction. These findings suggest a predisposition for liver damage and dysfunction in DM1 patients; however, this possibility has gone uninvestigated.
To understand the effects of DM1 in the liver, we generated a hepatocyte-specific DM1 mouse model in which we can induce the expression of CUG containing RNA, specifically in the liver. Through these mice, we have shown the presence of the toxic RNA in hepatocytes sequesters Mbnl proteins, causing a reduction in mature hepatocellular activity. We have characterized the transcriptomic changes driven by DM1 in the liver and have shown that these lead to changes in liver morphology, inflammation, necrosis, and increased lipid accumulation. We have demonstrated that DM1 sensitizes the liver to poor diet, increasing the likelihood of NAFLD development when patients consume high fat, high sugar diets. We have utilized the mouse models to demonstrate that DM1 adversely affects drug metabolism, slowing drug clearance and making patients more sensitive to potentially lethal anesthetics and analgesics. Finally, we have shown that divergence from appropriate adult hepatocellular function caused by DM1 increases with age, like other tissues affected by DM1.

Keywords: Myotonic Dystrophy Type 1, Alternative Splicing , Liver

24. Role of ADR-1 in the pathogen response of C. elegans

Alfa Dhakal (Cell, Molecular and Cancer Biology, Indiana University, School of Medicine), Halle Stump (Cell, Molecular and Cancer Biology, Indiana University, School of Medicine), Heather A. Hundley (Cell, Molecular and Cancer Biology, Indiana University, School of Medicine)

Abstract not available online - please check the booklet.

25. RNA G-quadruplex behavior and observation of liquid-liquid phase separation under biologically relevant conditions.

Taylor M. Dickson (Department of Immunology and Infectious Disease, Pennsylvania State University, University Park, PA), Allison M. Williams ( Department of Biochemistry and Molecular Biology, Center for RNA Molecular Biology, Pennsylvania State University, University Park, PA), Philip C. Bevilacqua (Department of Chemistry, Department of Biochemistry and Molecular Biology, Center for RNA Molecular Biology, Pennsylvania State University, University Park, PA)

Abstract:
G-quadruplexes are stable guanine-rich secondary nucleic acid structures that exist in vitro. RNA G-quadruplexes, unlike the typical double stranded DNA counterpart, are dependent upon environmental conditions for their formation and stability. While many studies investigating G-quadruplex formation and stability focus on conditions outside the realm of biology, we explore biological RNA sequences under biologically representative conditions. Liquid-liquid phase separation has been observed under a confocal in the presence of an oncogene, ions, polyamines, and small molecules. The varying observation of droplets and aggregates suggests that the formation of guanine interactions within RNA sequences is environmentally driven. We hypothesize that droplet and aggregate interactions interfere with G-quadruplex formation and function within cellular processes. This research suggests the condition-specific nature of secondary nucleic acid structure formation and emphasizes the importance of expanding upon RNA G-quadruplex structure analysis.

Keywords: RNA, Q-quadruplex , liquid-liquid phase separation

26. iCodon: Harnessing mRNA stability to customize gene expression

Michay Diez (Stowers Institute for Medical Research), Santiago Gerardo Medina-Muoz (Stowers Institute for Medical Research), Luciana Andrea Castellano (Stowers Institute for Medical Research), Gabriel da Silva Pescador (Stowers Institute for Medical Research), Qiushuang Wu (Stowers Institute for Medical Research), Ariel Alejandro Bazzini (Stowers Institute for Medical Research)

Abstract:
The codon composition of messenger RNA (mRNA) imposes regulatory information that strongly affects mRNA stability and fine-tunes protein expression. Current codon optimization methods revolve around codon usage frequency, despite the fact that it weakly correlates with stability. Here, we trained a machine learning model to predict mRNA stability based on the regulatory properties of codon composition. Using this model, we developed www.iCodon.org, a web interface that predicts stability, and customizes gene expression for vertebrate systems by introducing synonymous codon substitutions. To validate the potential of iCodon, we constructed twelve EGFP variants ranging in levels of predicted mRNA stability. Transfection of these variants in human cells revealed that our stability predictions correlated with fluorescence intensity and captured a range of nearly 50-fold differences in gene expression. Additionally, zebrafish embryos injected with these EGFP variants recapitulated the human cells results, demonstrating that iCodon can also modulate gene expression in vivo. Next, we explored potential applications of iCodon for basic biological research. First, we revealed how the estimated stability of gene families can be used to predict gene function and evolution. For example, core circadian genes displayed a significantly lower stability score compared to the stability of the transcriptome. Second, we showed that for heterologous gene expression in vertebrate systems iCodon outperforms gene optimization based on codon usage frequency by at least 3-fold. Finally, we found that iCodon stabilization improves in vivo loss-of-function phenotype rescues in later developmental stages in zebrafish embryos. In conclusion, iCodon provides a powerful tool to interrogate mRNA stability and design strategies to modulate gene expression in vertebrates, for a wide range of applications for research, and for the potential optimization of RNA-based therapeutics and vaccines.

Keywords: codon optimization, mRNA stability, evolutionary algorithm

27. Investigating the inhibitory role of initiation factor eIF2A on translation in vitro

Daisy J. DiVita (Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University), Michael G. Kearse (Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University)

Abstract:
Canonical translation uses the heterotrimeric eukaryotic initiation factor 2 (eIF2) as the initiator tRNA carrier and as a central node of regulation. However, eIF2 was not the first initiator tRNA carrier identified in eukaryotes. eIF2A (a monomer that is non-homologous to eIF2) was the first initiator tRNA binding protein discovered, but its role in translation was overshadowed after eIF2 was identified. Recent reports have shown that eIF2A is required for cancer progression, is not part of the canonical set of initiation factors, displays uncharacteristic GTP-independence for tRNA binding (unlike other tRNA-binding translation factors), and is able to stimulate initiation at CUG codons using Leu-tRNA(CUG); however, the exact function of eIF2A remains unknown. To determine how eIF2A functions in translation initiation, we programed in vitro translation extracts with recombinant human eIF2A. Our preliminary data show a 30-fold decrease in reporter activity when recombinant eIF2A is present, suggesting eIF2A is an inhibitor of translation. Here we will pursue testing how eIF2A is inhibiting translation and whether this inhibition requires specific factors during certain stages of initiation. Together, these mechanistic insights will shed light on how non-canonical initiation factors are used and regulate eukaryotic translation.

Keywords: Translational control, ribosome, protein synthesis

28. Robust methods to isolate, identify, and quantify tRNA modifications in prokaryotic and eukaryotic models

Taylor A. Dodson (Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo), Eric A. Carlson (Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo), Nathan C. Wamer (Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo), Chase N. Morse (Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo), Chao Liu, Samuel E. Senyo (Biomedical Engineering, School of Engineering, Case Western Reserve University ), Erin G. Prestwich (Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo)

Abstract not available online - please check the booklet.

29. Towards Understanding of the Role of Circular RNAs circ5615 and circCcnb1 in Cancerogenesis

Zihan Dong (Department of Chemistry, Mellon College of Science, Carnegie Mellon University), James M. Shockley (Department of Chemistry, Mellon College of Science, Carnegie Mellon University), Anna M. Kietrys (Department of Chemistry, Mellon College of Science, Carnegie Mellon University)

Abstract:
Circular RNAs (circRNAs) are a novel class of RNA molecules that are characterized by the covalent bond between the 3’ and 5’ ends generated through back-splicing 1. Various circular RNAs have the ability to sponge microRNAs (miRNAs) and proteins, affecting their cellular activities and leading to disease development and progression 1. Our project focuses on two circRNAs that have contrasting effects on carcinogenesis. First, a hsa_circ_0005615 (circ5615) has been linked to colorectal cancer progression when overexpressed. Circ5615 sponges miR-149-5p, and subsequently causes overexpression of the TNKS gene and upregulation of the cyclin D1 gene (CCND1) 2. In contrast, it has been found that expression of hsa_circ_0072758 (circCcnb1) helps to slow down the progression of breast cancer. This circRNA binds both Cyclin B1 and Cyclin Dependent Kinase 1(Cdk1) proteins, inhibiting formation of Cdk1-Cyclin B1 complex 3. CircCcnb1 sponge-like activity decreases cell proliferation and ultimately the progression of breast cancer in mice 3. However, how circ5615 and circCcnb1 interact with various proteins and miRNAs, and why the linear forms of mRNA cannot have the same inhibitory effect has not been studied.
In our work, we focus on the structural aspects of circRNAs to understand how the RNA structure may determine their sponge-like activity towards proteins and miRNAs. To map secondary structure of circRNAs, and mRNAs of their origin, we apply bioinformatic methods and in vivo selective 2’-hydroxyl acylation analyzed by primer extension (SHAPE). The difference between each circRNA’s structure and its linear counterpart provides insight into the various interactions possible for each circRNA and helps to explain its mode of action. Further, we localize sequential and structural binding motifs involved in interactions with proteins and miRNAs. These motifs may be further used to analyze the regulatory roles of other circRNAs, hopefully providing new targets for therapeutic treatment and early diagnosis.

References:
(1) Lee, E. C. S.; Elhassan, S. A. M.; Lim, G. P. L.; Kok, W. H.; Tan, S. W.; Leong, E. N.; Tan, S. H.; Chan, E. W. L.; Bhattamisra, S. K.; Rajendran, R.; Candasamy, M. The Roles of Circular RNAs in Human Development and Diseases. Biomedicine and Pharmacotherapy. Elsevier Masson SAS March 1, 2019, pp 198–208. https://doi.org/10.1016/j.biopha.2018.12.052.

(2) Ma, Z., Han, C., Zia, W. et al. circ5615 functions as a ceRNA to promote colorectal cancer progression by upregulating TNKS. Cell Death Dis 11, 356 (2020) http://doi.org/10.1038/s41419-020-2514-0

(3) Fang, L.; Du, W. W.; Awan, F. M.; Dong, J.; Yang, B. B. The Circular RNA Circ-Ccnb1 Dissociates Ccnb1/Cdk1 Complex Suppressing Cell Invasion and Tumorigenesis. Cancer Lett. 2019, 459 (May), 216–226. https://doi.org/10.1016/j.canlet.2019.05.036.

Keywords: circular RNA, cancerogenesis, RNA structure and function

30. Structural and functional characterization of Hfq C-terminal domain using native mass spectrometry

Chen Du (Resource for Native Mass Spectrometry Guided Structural Biology; The Department of Chemistry and Biochemistry, The Ohio State University), Mengxuan Jia (Resource for Native Mass Spectrometry Guided Structural Biology; The Department of Chemistry and Biochemistry, The Ohio State University), Samantha H. Sarni (Resource for Native Mass Spectrometry Guided Structural Biology; The Ohio State Biochemistry Program, The Ohio State University), Jorethe Roca, Ewelina Malecka-Grajek (T. C. Jenkins Department of Biophysics, Johns Hopkins University), Sarah Woodson (T. C. Jenkins Department of Biophysics, Johns Hopkins University), Vicki H. Wysocki (Resource for Native Mass Spectrometry Guided Structural Biology; The Department of Chemistry and Biochemistry, The Ohio State University; The Ohio State Biochemistry Program, The Ohio State University)

Abstract not available online - please check the booklet.

31. KREH1 helicase activity promotes the occurrence and proper positioning of mRNA editing initiation in Trypanosoma brucei

Ashutosh P Dubey (Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA), Brianna L. Tylec (Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA), Amartya Mishra (Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA), Runpu Chen (Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA), Yijun Sun (Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA), Laurie K. Read (Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA)

Abstract:
In trypanosomes, 12 of 18 mitochondrially encoded mRNAs require post-transcriptional insertion and deletion of uridine residues to create translatable mRNAs. Editing is directed by sequentially utilized trans-acting guide RNAs (gRNAs) that base pair with mRNA, and catalyzed by a multi-subunit holoenzyme. The ATP-dependent RNA helicase, Kinetoplast RNA Editing Helicase 1 (KREH1), interacts transiently with the editing holoenzyme and was previously suggested to be involved in the removal of gRNAs from mRNAs following their utilization. To further investigate the KREH1 function, we generated a KREH1 null mutant (KO) in procyclic form T. brucei. KREH1 KO exhibits a modest decrease in a subset of edited mRNAs compared to wild type (WT), in particular ATPase (A6) mRNA. High throughput sequence analysis of A6 mRNA in KREH1 KO cells revealed no evidence for KREH1 function in gRNA removal. Rather, we identified multiple sequences containing a long stretch of pre-edited sequence followed by one or two modified editing sites outside of the region whose editing is directed by the first gRNA. To define the role of KREH1 helicase activity, as opposed to simply its presence, we overexpressed WT KREH1 or KREH1 with mutations in either in the conserved ATP-binding motif (KA) or DEAD box domain (EQ). Both the KA and EQ mutants, but not the WT overexpressors, exhibit a dominant-negative (DN) phenotype marked by a dramatic decrease initiation of editing on most mRNAs with concomitant accumulation of pre-edited transcripts, and defects in the positioning of the initial A6 mRNA editing event similar to those observed in KO cells. Despite the differing effects of WT and KA KREH1 on editing initiation, co-IP and RIP analyses show that these proteins interact similarly with the editing holoenzyme, mRNA, and gRNA. Together, our data support a model in which KREH1 helicase activity modulates RNA-RNA interactions to promote the occurrence and proper positioning of editing initiation.

Keywords: Trypansoma, RNA Helicase, RNA editing

32. Investigating the role of EFTUD2 in pre-mRNA splicing and EJC deposition

Caleb M. Embree (Department of Molecular Genetics, Center for RNA Biology), Guramrit Singh (Department of Molecular Genetics, Center for RNA Biology)

Abstract:
During the gene expression pathway, pre-mRNA splicing is one of the earliest and most important RNA processing steps, which occurs co-transcriptionally. The end result of pre-mRNA splicing is an mRNP particle comprised of mRNA and RNA binding proteins deposited during splicing. One complex of RNA binding proteins, the exon junction complex (EJC), plays important roles in steps ranging from pre-mRNA splicing in the nucleus to mRNA degradation in the cytoplasm. Though the EJC is deposited by the spliceosome, the responsible spliceosomal proteins and their precise function in EJC deposition is not fully known. EFTUD2, a splicing regulator and member of the U5 snRNP that sits adjacent to the EJC within the activated spliceosome, may have a role in EJC deposition. Mutations in EFTUD2 and the EJC core factor EIF4A3 cause similar cranio-facial disorders, hinting that they function in a similar pathway. Using clinically relevant C-terminally truncated EFTUD2 proteins we are investigating the relationship between EFTUD2 and the EJC. Our work shows that the C-terminal truncations of EFTUD2 destabilize the protein, which suggests that these mutations likely cause EFTUD2 haploinsufficiency in patients. Our hypothesis is that such a haploinsufficiency leads to defects in spliceosome and/or EJC assembly resulting in splicing errors. Using EFTUD2 haploinsufficient (or knockdown) HEK293 cells, we plan to identify the splicing errors via RNA-Seq and assess transcriptome-wide spliceosome assembly via spliceosome profiling. Together these data will allow us to determine how reduced levels of EFTUD2 impairs spliceosome and/or EJC assembly leading to splicing defects.

Keywords: spliceosome, EFTUD2

33. Investigating an ADAR-dependent germline phenotype in 𝘊𝘢𝘦𝘯𝘰𝘳𝘩𝘢𝘣𝘥𝘪𝘵𝘪𝘴 𝘦𝘭𝘦𝘨𝘢𝘯𝘴

Emily A. Erdmann (Department of Biology, Indiana University ), Melanie Forbes, Maggie Holdeman, Olivia Abraham, Heather A. Hundley (Medical Sciences Program, Indiana University School of Medicine- Bloomington)

Abstract not available online - please check the booklet.

34. Computational discovery of novel functional RNAs in SARS-CoV-2

Peter C. Forstmeier (Department of Biochemistry and Molecular Biology - The Pennsylvania State University), McCauley O. Meyer (Department of Biochemistry and Molecular Biology - The Pennsylvania State University), Philip C. Bevilacqua (Pennsylvania State University, Department of Biochemistry and Molecular Biology, Department of Chemistry)

Abstract:
The global COVID-19 pandemic is caused by the positive-sense RNA virus SARS-CoV-2 (1). The genome of this virus is known to hold functional RNAs, a type of biologically essential RNA characterized by conserved sequence, conserved structure, and presence of a pseudoknot (2). Functional RNAs are also good therapeutic targets for antisense oligonucleotides (ASOs) or small molecules making the discovery of novel functional RNAs in SARS-CoV-2 a route for developing therapeutics to counter the disease (3). We built a computational pipeline that functions in two parts. The first part agnostically locates structured RNAs in the genome and determines which of those structures contain pseudoknots; those that contain pseudoknots continue through the pipeline. This part also eliminates terminators and known riboswitches and ribozymes. The second part of the pipeline uses thousands of viral genomic isolates to determine conservation of sequence, structure, and pseudoknots. A known functional RNA within the SARS-CoV-2 genome is a programmed (–1) ribosomal frameshifting element (FSE), which was also analyzed using the pipeline to establish a benchmark for a level of mutation that was acceptable (4). Pseudoknots that showed higher levels of conservation of sequence, secondary structure, and pseudoknot structure than the FSE were considered putatively functional. A final level of analysis was added to the pipeline that eliminated mutations that cause nonsense or missense non-conservative amino acid mutations. The rationale being that it is ambiguous whether the selective pressure acting on those mutations is due to RNA structure or the protein produced. Using this pipeline, seven putatively functional RNAs were identified in the SARS-CoV-2 genome that contain highly conserved sequence, secondary structure, and a pseudoknot. These RNAs are potential novel therapeutic targets.

References:
(1) Manfredonia, I.; Nithin, C.; Ponce-Salvatierra, A.; Ghosh, P.; Wirecki, T. K.; Marinus, T.; Ogando, N. S.; Snijder, E. J.; Hemert, M. J. van; Bujnicki, J. M.; Incarnato, D. Genome-Wide Mapping of Therapeutically-Relevant SARS-CoV-2 RNA Structures. bioRxiv 2020.
(2) Peselis, A.; Serganov, A. Structure and Function of Pseudoknots Involved in Gene Expression Control. Wiley interdisciplinary reviews. RNA. NIH Public Access November 1, 2014, pp 803–822.
(3) Crooke, S. T.; Witztum, J. L.; Bennett, C. F.; Baker, B. F. RNA-Targeted Therapeutics. Cell Metabolism. Cell Press April 3, 2018, pp 714–739.
(4) Sun, Y.; Abriola, L.; Surovtseva, Y. V; Lindenbach, B. D.; Guo, J. U. Restriction of SARS-CoV-2 Replication by Targeting Programmed −1 Ribosomal Frameshifting In Vitro. bioRxiv 2020.

Keywords: SARS-CoV-2, RNA, Pseudoknots

35. 5-methyluridine modifications in mRNA codons impact protein synthesis by the ribosome

Monika K Franco (University of Michigan Program in Chemical Biology), Joshua D Jones (University of Michigan, Department of Chemistry), Mehmet Tardu (University of Michigan, Department of Chemistry), Laura R Snyder (University of Michigan, Department of Chemistry), Robert T Kennedy (University of Michigan, Department of Chemistry), Kristin S Koutmou (University of Michigan Program in Chemical Biology, University of Michigan, Department of Chemistry)

Abstract not available online - please check the booklet.

36. RNA Pseudoknot Structure in 16S Helix 18 Destabilizes the Binding and Enzymatic Activity of Ribosomal RNA Modification Enzyme RsuA

Sanjaya Abeysirigunawardena (Kent State University), Kumudie Jayalath (Kent State University), Sean Frisbie (Kent State University )

Abstract:
Ribosomes are ribonucleoprotein complexes that are involved in protein
biosynthesis in cells. Ribosomes are generated from the assembly of
three different ribosomal RNAs (rRNAs) and more than 50 ribosomal
proteins (r-proteins). Though the ribosome assembly is well studied, the
impact of post-transcriptional rRNA modification enzymes and their
respective modifications on ribosomal assembly is not completely
understood. In response to the lack of understanding of preferred
substrates and the structural prerequisites for the stable binding and
function of the rRNA modification enzymes, we study thermodynamics
of protein RsuA to rRNA. My study focused on understanding the impact
of pseudoknot structure of 16S helix 18 on the binding of RsuA. Several
rRNA mutants that are incapable of forming 16S helix 18 pseudoknot
was generated by using site-directed mutagenesis. FRET-based binding
assay are used to measure the thermodynamic stability of RsuA-rRNA
complexes. These findings will enable us to elucidate thermodynamically
preferred mechanism of RsuA binding to rRNA.

References:
https://www.mdpi.com/2218-273X/10/6/841

Keywords: Helix 18, 16s RNA, Enzymatic Activity

38. Probing the impact of HOPS on RNAs with single molecule tracking

Guoming Gao (Biophysics Graduate Program and Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA), Nils G Walter (Department of Chemistry, Biophysics, and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA)

Abstract:
Proteins and RNAs can form functional biological condensates, also known as membraneless organelles, via liquid-liquid phase separation (LLPS). The partitioning of different proteins and RNAs between the dilute phase and the condensed phase provides delicate regulation over their functions, from promoting biochemical reactions and specific intermolecular interactions, to sequestering key molecules from downstream processing or signaling. Hyperosmotic phase separation (HOPS) is a recently discovered LLPS triggered by the osmotic compression of cell volume. A majority of homo-multimeric proteins are shown to undergo HOPS in several cell types. Moreover, HOPS is much faster than most cellular LLPS processes (within ~10 s versus over minutes to hours), and thus HOPS condensates could be first responders sensing cell volume change and priming other stress responses. However, it was unclear whether RNAs contribute to HOPS and how HOPS impacts the diffusion behaviors and functions of different RNAs. Here, we used live-cell single molecule RNA tracking to explore the dwelling of mRNAs, lncRNAs, and miRNAs in HOPS condensates, and measure the change in their diffusion behaviors in the presence of HOPS. A theoretical model for RNA’s role in HOPS is proposed based on RNA interactions with enigmatic RNA binding sites (EnigmRBS) on protein surfaces.

Keywords: LLPS, Single Molecule

39. The p38 MAPK Hog1 remodels the translatome of the fungal pathogen Cryptococcus neoformans.

David Goich (Microbiology and Immunology, University at Buffalo), Amanda L. M. Bloom (Microbiology and Immunology, University at Buffalo), John C. Panepinto (Microbiology and Immunology, University at Buffalo)

Abstract:
Cryptococcus neoformans is an environmental fungus that causes severe opportunistic infections in immunocompromised individuals, particularly people living with HIV/AIDS. Upon entry into the host, C. neoformans rapidly alters its proteome to accommodate a variety of stressors, including elevated temperature, reactive oxygen species (ROS) in phagocytes, nutrient deprivation, and more. Our preliminary data indicate that these processes are dependent in part on the p38 MAPK, Hog1, which couples the translational response to stress-sensing signal transduction. Using polysome profiling, translational output assays, and mRNA repression kinetics, we investigated whether two putative kinases downstream of Hog1, Hrk1 and Cmk2, are similarly defective in translatome reprogramming. We also investigated the role of these proteins in the translational response to fludioxonil, an antifungal with Hog1-dependent activity. We found that while hrk1∆ demonstated delayed recovery from translation repression, deletion of Cmk2 caused dysregulation of eIF2α. Our data suggests that both of these putative effectors of Hog1 play a role in translational adaptation to stress. Ongoing investigations will determine if these proteins play stress-specific roles, as well as the role of each in the translational response to fludioxonil.

Keywords: Translation

40. The novel role of m6A-mRNA binding protein YTHDF1 in the regulation of heart function

Volha A Golubeva (Medical Scientist Training Program and Biomedical Sciences Graduate Program, Center for RNA Biology, OSU, Columbus OH), Lisa Dorn (Medical Scientist Training Program, Center for RNA Biology, OSU, Columbus OH), Federica Accornero (Center for RNA Biology, Davis Heart and Lung Research Institute and Department of Physiology and Cell Biology, OSU, Columbus OH)

Abstract not available online - please check the booklet.

41. Alternative polyadenylation of transcripts encoding trans-acting siRNAs in red clover

Caleb Gooden (Department of Plant and Soil Sciences, University of Kentucky), Randy D. Dinkins (USDA-ARS-FAPRU), Arthur G. Hunt (Department of Plant and Soil Sciences, University of Kentucky)

Abstract:
Root nodulation is a process in which host roots are infected with a soil bacterium and form nodules that better fix nitrogen from the atmosphere. In agriculture, nodules help to reduce the need for fertilizer and are thus part of sustainable practice. Studies in Medicago truncatula [1] have shown the process of root nodulation to be suppressed by the expression of a regulatory long non-coding RNA (TAS3). Over-expression of TAS3 and the miR390 pathway with which it interacts resulted in prioritization of lateral root formation over nodulation. Thus, TAS3 is an attractive target for manipulation to improve nitrogen fixation in legumes and enhance their agronomic value.

In Arabidopsis, TAS3a transcripts may end at one of two alternative polyadenylation sites, the choice of which determines whether functional or non-functional TAS3a RNAs are made [2]. Specifically, RNAs ending at the promoter-proximal site will lack the characteristic miR390 target site and thus be non-functional. TAS3a transcript ending at the distal site will possess the miR390 target site, and thus can serve as substrates for miR390-directed cleavage and subsequent production of siRNAs that regulate the responses to auxin.

We hypothesized that if two polyadenylation (poly(A)) sites are present in the TAS3 region of Trifolium pratense (red clover), then differential use of these sites would be seen in lateral roots and nodules. To test whether red clover TAS3 transcripts end at different poly(A) sites, we utilized 3’-RACE to identify and sequence TAS3 RNA isoforms. The results confirm that transcripts encoding the red clover TAS3 may end at one of two poly(A) sites. Further bioinformatics analysis showed that a miR390 target sequence is situated between the two poly(A) sites. These results confirm that red clover TAS3 transcripts undergo alternative polyadenylation much as is seen in Arabidopsis and M. truncatula, and that alternative polyadenylation may regulate TAS3 function in red clover. Current research focuses on measuring the differential use of these sites in roots and nodules using quantitative PCR (qPCR) and 3’-RACE assays. Using these results as a guide, we plan on developing transgenic approaches to manipulate the relative usages of these sites to further establish their importance on nodulation.

References:
1. Hobecker KV, Reynoso MA, Bustos-Sanmamed P, Wen J, Mysore KS, Crespi M, et al. The MicroRNA390/TAS3 Pathway Mediates Symbiotic Nodulation and Lateral Root Growth. Plant Physiol. 2017;174(4):2469-86.
2. Hunt AG. RNA regulatory elements and polyadenylation in plants. Front Plant Sci. 2011;2:109.

Keywords: alternative polyadenylation, siRNA, nodulation

42. Gene-specific Pulldown for targeted in vivo RNA structure probing

Samantha Grecco (Department of Biochemistry and Molecular Biology, Beckman Scholar, Schreyer Honors College, Pennsylvania State University, University Park, Pennsylvania, USA), Janie K. Frandsen (Department of Chemistry, Center for RNA Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA), Philip C. Bevilacqua (Department of Biochemistry and Molecular Biology, Department of Chemistry, Center for RNA Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA)

Abstract:
When Escherichia coli experiences environmental stressors it initiates regulatory responses to maintain homeostasis. In response to a specific stress, small non-coding RNAs (sRNAs) change the expression of target mRNAs by altering mRNA stability and translation efficiency. The sRNA RyhB is expressed under iron-limiting conditions and regulates the iron sparing response.1 The majority of RyhB targets have not been characterized and the role of RNA structure in the sRNA-mRNA interactions is unknown. It is hypothesized that the target mRNA secondary structure influences the ability of RhyB to bind and that the interaction may induce a structural rearrangement to alter gene expression. To analyze the role of RNA structure in RyhB regulation four putative RyhB targets (cirA, fliA, sufAB, frdA) were selected. Then gene expression profiling was performed, and a gene-specific pulldown was developed for targeted in vivo structure probing. This research will provide a deeper understanding of the regulatory systems in E. coli, which could help combat pathogenic infections and antibiotic resistance.

References:
1. Chareyre, S., Mandin, P. (2018) Bacterial Iron Homeostasis Regulation by sRNAs, Microbiol. Spectr. 6(2), 267-281.

Keywords: sRNA, RyhB, RNA structure probing

43. Translation reinitiation regulates oocyte maturation

Lydia Grmai (Department of Cell Biology, University of Pittsburgh), Hyung Don Ryoo (Department of Cell Biology, New York University School of Medicine), Deepika Vasudevan (Department of Cell Biology, University of Pittsburgh)

Abstract not available online - please check the booklet.

44. Characterization of putative SAM-I riboswitches of L. monocytogenes

Ian Hall (Department of Chemistry, University of Michigan), Sarah Keane (Department of Chemistry and Program in Biophysics, University of Michigan)

Abstract:
Listeria monocytogenes is a saprophytic bacterium which can infect humans causing listeriosis, a life-threatening foodborne illness. In the U.S. about 2,500 cases of listeriosis and 500 related deaths are reported each year. Understanding the bacteria’s virulence and metabolism are essential for improving strategies to prevent and treat listeriosis. Notably regulation of the bacteria’s virulence gene expression is primarily controlled by positive regulatory factor A (PrfA) and an RNA thermosensor (RNAT) that resides in the 5′ untranslated region (UTR) of the prfA messenger (m)RNA. We previously described the mechanism of the prfA RNAT’s temperature dependent translation regulation.1 PrfA translation research revealed a novel trans interaction between the prfA RNAT and S-adenosyl methionine (SAM) response element A (SreA)2, a putative SAM-I riboswitch identified in tiling arrays of the L. monocytogenes genome.3
The present work biophysically characterizes SreA and six other putative SAM-I riboswitches of L. monocytogenes (SreA-G). These elements reside in front of genes for SAM biosynthesis pathways and/or SAM related transport pathways. A key hypothesis of this work is whether the genomic location of a riboswitch is related to its regulatory strength. To answer this question and characterize these presumed SAM-I riboswitches, we initiated studies on the ligand binding, transcription termination, and structure of SreA-G. The ligand affinity and specificity of SreA-G are being determined by isothermal titration calorimetry (ITC) using cognate (SAM) and near-cognate (SAH) ligands. The transcription termination of the riboswitches with SAM or SAH are being evaluated by a 32P GTP body labeled assay. The aptamer domains of the riboswitches were studied by small angle X-ray scattering (SAXS) with and without SAM, and in all cases, we observe compaction of the structure with SAM. Additional structural studies of aptamer domains via X-ray crystallography are underway.

References:
1. Zhang, H.; Hall, I.; Nissley, A. J.; Abdallah, K.; Keane, S. C., A Tale of Two Transitions: The Unfolding Mechanism of the prfA RNA Thermosensor. Biochemistry 2020, 59 (48), 4533-4545.
2. Loh, E.; Dussurget, O.; Gripenland, J.; Vaitkevicius, K.; Tiensuu, T.; Mandin, P.; Repoila, F.; Buchrieser, C.; Cossart, P.; Johansson, J., A trans-acting riboswitch controls expression of the virulence regulator PrfA in Listeria monocytogenes. Cell 2009, 139 (4), 770-9.
3. Toledo-Arana, A.; Dussurget, O.; Nikitas, G.; Sesto, N.; Guet-Revillet, H.; Balestrino, D.; Loh, E.; Gripenland, J.; Tiensuu, T.; Vaitkevicius, K.; Barthelemy, M.; Vergassola, M.; Nahori, M.-A.; Soubigou, G.; Régnault, B.; Coppée, J.-Y.; Lecuit, M.; Johansson, J.; Cossart, P., The Listeria transcriptional landscape from saprophytism to virulence. Nature 2009, 459 (7249), 950-956.

Keywords: SAM-I riboswitch, Listeria monocytogenes

45. Establishing the presence of hidden breaks in the chloroplast rRNA of various plants

Cooper Halliday (University of Pittsburgh at Johnstown), Kaylin Gaudette (University of Pittsburgh at Johnstown), Laura Ritchey (University of Pittsburgh at Johnstown)

Abstract:
In plant cells, light is captured in specialized organelles known as chloroplasts. These structures turn light energy into chemical energy in the process of photosynthesis. Like mitochondria, chloroplasts contain their own DNA as well as their rRNA. It was discovered that there are two hidden breaks in the rRNA of the 23S large ribosomal subunit. (1-3) The existence of hidden breaks has been detected in the rRNAs of plants, insects (in the 28S rRNA), and in rodents. (1) These breaks, in plants, are believed to be important for translation within the chloroplast and are the result of specific nuclease-mediated (1,2) covalent breaks in the backbone of the rRNA. We have hypothesized that stress conditions such as cold and hot temperatures affect the expression of these breaks. We extracted RNA from Oryza sativa and Spinacia oleracea subjected to temperature stress conditions and analyzed the RNA using qualitative assays to identify the presence of the hidden breaks. We also aim to use sequencing gels and RT-qPCR to look at the nucleotide-specific location in the sequence that the hidden break occurs in the rRNA for each of the plants. Once grown, Coffea arabica and Arabidopsis thaliana will be run through the same procedures to have a wider organism pool for comparison.

References:
1. Nishimura, K., et al. Plant J 63 (5), 766-77 (2010).
2. Liu, J., et al. Plant Physiol 168 (1), 205-221, (2015).
3. Ritchey, L.E., et al. Nucleic Acids Res 45 (14), el35 (2017).

Keywords:

46. Modulating RNA Splicing of DNA Topoisomerase IIα in Human Leukemia K562 Cells: Use of CRISPR/Cas9 Gene Editing to Impact Sensitivity/Resistance to the Anticancer Agent Etoposide

Victor A. Hernandez (Pharmaceutics and Pharmacology, The Ohio State University ), Jessika Carvajal (Pharmaceutics and Pharmacology, The Ohio State University ), Jonathan L. Papa, Nicholas Shkolnikov (Pharmaceutics and Pharmacology, The Ohio State University ), Hatice Gulcin Ozer (Biomedical Informatics, The Ohio State University ), Jack C. Yalowich, Terry S. Elton (Pharmaceutics and Pharmacology, The Ohio State University ), Junan Li (Pharmaceutics and Pharmacology, The Ohio State University )

Abstract:
The human DNA topoisomerase IIα (170 kDa, TOP2α/170) is an essential enzyme that resolves DNA topological entanglements that form during chromosome condensation and segregation, making it indispensable for the survival of highly proliferating cells including cancer cells. Hence, TOP2α is a prominent target for anticancer therapies. Some of the most widely used TOP2 targeting drugs such as etoposide, stabilize a TOP2α/170-DNA cleavage complex preventing the religation of broken DNA strands and exerting cytotoxic effects by the accumulation of DNA damage.
Acquired chemoresistance to TOP2 targeting drugs continues to be a major obstacle in cancer treatment in the clinic. Our lab developed etoposide resistant human leukemia K562 cells, designated K/VP.5 in which levels of TOP2α/170 were decreased along with identification of a novel C-terminal truncated isoform of TOP2α, TOP2α/90. TOP2α/90 is the translation product of novel alternatively spliced mRNA via intron 19 (I19) retention and processing. TOP2α/90 is a determinant of acquired resistance to etoposide through a dominant-negative effect related to heterodimerization with TOP2α/170.
It was hypothesized that the E19/I19 splice site (SS) is inefficiently recognized by the spliceosome and results in decreased levels of TOP2α/170 and drug resistance. Therefore, CRISPR/Cas9 was utilized for gene editing of TOP2α in K/VP.5 cells to enhance I19 removal and circumvent acquired TOP2α-mediated drug resistance. Gene edited clones were identified by qPCR and verified by sequencing. Characterization of a K/VP.5 clone with all TOP2α alleles edited revealed improved I19 removal, decreased TOP2α/90 protein, and increased TOP2α/170 protein. In addition, gene editing to optimize the TOP2α E19/I19 SS in K/VP.5 cells circumvented resistance to TOP2α-targeted agents.
Conversely, experiments were performed whereby the E19/I19 SS in etoposide sensitive K562 cells was gene edited to decrease/eliminate splicing at this boundary. Inhibiting splicing of the E19/I19 SS in K562 cells resulted in a decrease in full-length TOP2α/170 expression and induction of resistance to etoposide. Together, results support the role of alternative splicing at the E19/I19 boundary as a determinant of resistance to TOP2α-targeting agents.

Keywords: CRISPRCas9, Alternative RNA Splicing , Drug Resistance

47. Strand Invasion during the Fluoride Riboswitch Cotranscriptional Folding Pathway

Laura M Hertz (Department of Chemical and Biological Engineering at Northwestern University), Angela M Yu (Electrical and Computer Engineering Department at University of Washington), Julius B Lucks (Department of Chemical and Biological Engineering at Northwestern University)

Abstract not available online - please check the booklet.

49. Stress-Induced Increase in Small ncRNAs Generated from Regulatory Regions of the Mitochondrial Genome

Kathryn E. Holzmacher (Department of Biochemistry at University at Buffalo), Jonathan E. Bard (Center of Excellence in Bioinformatics and Life Sciences, and Department of Biochemistry at University at Buffalo), Theodore L. Mathuram (Department of Biochemistry at University at Buffalo), Natalie A. Lamb (Center of Excellence in Bioinformatics and Life Sciences at the University at Buffalo), Norma J. Nowak (Center of Excellence in Bioinformatics and Life Sciences, and Department of Biochemistry at University at Buffalo), A. Blumental-Perry (Department of Biochemistry at University at Buffalo)

Abstract:
Rationale: Alveolar Epithelial Type (AET) 2 cells serve as progenitors for the repair of destructed alveoli. We identified AET2-specific mitochondria-associated retrograde signaling via mtDNA-encoded non-coding (nc) RNA, mito-ncR-805. mito-ncR-805 levels are increased in the mitochondria and nucleus during stress, and positively regulate mitochondrial metabolism and respiration. We propose that mito-ncR-805 represents a subtype of mitochondrial small RNAs that protect cells under stress. To identify transcripts with similar functions we compared mitochondrial transcriptomes of unstressed and stressed cells.
Methods: CS-exposures of MLE12 cells were used to study adaptational transcriptomes. Stress signaling was evaluated by Western blot. Mitochondrial small RNA transcriptome was evaluated by constructing and analyzing strand-specific RNAseq libraries of transcripts 100 bp and shorter. A bioinformatic pipeline was specifically developed for the analysis of mitochondrial transcripts. Identified transcripts were validated by Northern blot and qPCR analyses.
Results: CS exposure increased the generation of 3 small RNAs. The transcripts had a specific processing pattern and originated from the control regions of the mitochondrial genome (D-loop and Rnr genes). The presence of the transcripts in cells and their elevation during smoke exposure was confirmed by Northern. Mitochondrial origin was validated in either mitochondrial-depleted cells (grown in EtBr), or in cells where mitochondrial transcription was inhibited by 6G-rhodamine. Cellular localization of Rnr-derived transcripts is currently being evaluated.
Conclusion: We demonstrate the stress-dependent generation of 4 distinct transcripts that originate from control regions of the mitochondrial genome. This suggests a possible regulatory function in coordinating the activities in both mitochondrial and nuclear genomes. We propose that those small RNAs aid in signaling which aim to restore mitochondrial homeostasis.

References:
Blumental-Perry A, Jobava R, Bederman I, Degar AJ, Kenche H, Guan BJ, Pandit K, Perry NA, Molyneaux ND, Wu J, Prendergas E, Ye ZW, Zhang J, Nelson CE, Ahangari F, Krokowski D, Guttentag SH, Linden PA, Townsend DM, Miron A, Kang MJ, Kaminski N, Perry Y, Hatzoglou M. Retrograde signaling by a mtDNA-encoded non-coding RNA preserves mitochondrial bioenergetics. Commun Biol. 2020 Oct 30;3(1):626. doi: 10.1038/s42003-020-01322-4. PMID: 33127975; PMCID: PMC7603330.
Lee C, Zeng J, Drew BG, Sallam T, Martin-Montalvo A, Wan J, Kim SJ, Mehta H, Hevener AL, de Cabo R, Cohen P. The mitochondrial-derived peptide MOTS-c promotes metabolic homeostasis and reduces obesity and insulin resistance. Cell Metab. 2015 Mar 3;21(3):443-54. doi: 10.1016/j.cmet.2015.02.009. PMID: 25738459; PMCID: PMC4350682.

Keywords: Alveolar Epithelial Type 2 Cells, Mitochondrial Stress Signaling , Small non-coding RNA

50. RNA Thermometer Thermal Stability Modulated by pH-Sensitive Molecular Switching

Md Ismail Hossain (Department and institution: Department of Chemistry & Biochemistry, Ohio University, Athens, OH), Erin R. Murphy (Department and institution: Department of Biomedical Sciences, Ohio University Athens, OH), Jennifer V. Hines (Department and institution: Department of Chemistry & Biochemistry, Ohio University, Athens, OH)

Abstract:
Bacterial resistance to current medicines continues to be a significant global health threat. Consequently, there is a need to develop new therapeutic approaches that prevent or overcome bacterial infections. One emerging area of investigation is targeting regulatory RNA, which mediates gene expression in bacteria. In order for drug discovery studies to be successful, a detailed understanding of the molecular mechanism of the RNA-mediated regulation is needed. In this study, we investigated the effect of pH on the thermal stability of a set of RNA thermometers (RNATs) found in the untranslated leader region of Shigella shuA, shuT, and ompA mRNA. 1,2 Shu (Shigella heme utilization) proteins have essential roles in utilizing heme as a sole source of iron. Outer membrane protein A (ompA) is a virulence factor required for the cell adhesion, biofilm formation, invasion, and spread of the bacteria. UV monitored thermal denaturation was used to determine the melting temperature (Tm) and thermodynamic parameters for model RNAs of each RNAT. At low ionic strength, the Tm of the shuT and ompA RNA thermometers were pH dependent. However, at high ionic strength, only the Tm of shuT was pH dependent. The Tm of shuA was not pH-dependent at either low and high ionic strength, but it was significantly higher than that of shuT and ompA. We also investigated the effect of pH on RNAT binding of a 16S rRNA mimic using a fluorophore-quencher labeled model RNA. For ompA, the EC50 for binding the 16S rRNA mimic was pH dependent. These results indicate that in some RNA thermometers, pH-sensitive molecular switching may play a role in modulating gene expression regulation.

References:
1. Kouse, Andrew B., Francesco Righetti, Jens Kortmann, Franz Narberhaus, and Erin R. Murphy. "RNA-mediated thermoregulation of iron-acquisition genes in Shigella dysenteriae and pathogenic Escherichia coli." PloS one 8, no. 5 (2013): e63781.
2. Murphy, Erin R., Johanna Roßmanith, Jacob Sieg, Megan E. Fris, Hebaallaha Hussein, Andrew B. Kouse, Kevin Gross et al. "Regulation of OmpA translation and Shigella dysenteriae virulence by an RNA thermometer." Infection and immunity 88, no. 3 (2020): e00871-19.

Keywords: RNA Thermometer, shuT, shuA, OmpA, Molecular switching

51. Determining the role of the 3’-5’ reverse polymerase in Myxococcus xanthus

Brandon Iwaniec (Chemistry and Biochemistry, The Ohio State University), Jane Jackman (Chemistry and Biochemistry, The Ohio State University)

Abstract:
tRNAHis guanylyltransferase-like proteins (TLPs) are able to catalyze nucleotide addition to RNA substrates in the opposite direction (3'-5') of all known canonical RNA polymerases. This biochemical activity is seen in vitro with bacterial TLPs, which can add nucleotides to the 5'-ends of tRNA substrates. However, no bona fide biological substrates have been identified to date in any bacterial species that has a TLP. Myxococcus xanthus, a bacterium, exhibits defects in starvation-induced fruiting body formation and sporulation when its MXTLP gene is deleted, suggesting that this enzyme plays a role in maturation or maintenance of some biologically relevant RNA species. Here, we used a variety of in vitro biochemical assays to gain a more in-depth picture of the biological role of MxTLP in M. xanthus. Addition of an essential G-1 nucleotide to the 5'-end of tRNAHis is an important function of members of the tRNAHis guanylyltransferase (Thg1) enzyme family. This activity has also been associated with one TLP, and therefore we sought to test whether MxTLP could exhibit a similar function. However, we demonstrated that MxTLP is not apparently needed for addition of the G-1 nucleotide in vivo in M. xanthus, based on primer extension assays of RNA isolated from MXTLP deletion strains. This observation is also consistent with the RNase P-dependent pathway for obtaining G-1 in bacteria, and our in vitro histidylation assays confirmed that the form of G-1-containing tRNAHis preferred by the M. xanthus histidyl tRNA synthetase is the form that would most likely result from post-transcriptional RNase P cleavage, rather than from addition by MxTLP. We also demonstrated that MxTLP can incorporate labeled nucleotides into RNA isolated from vegetative and developing cells, but with different patterns of incorporation, indicating that MxTLP may play distinct roles in each life cycle. Efforts to identify the substrates that are acted on during each life cycle stage using pull-down approaches are underway, and these results will be used to provide the first look at the potential biological function of a TLP in any bacterial system.

Keywords: Reverse polymerization , tRNA, Myxococcus xanthus

52. Reverse (3'-5') RNA polymerases: Applications beyond tRNA(His) maturation

Malithi I. Jayasinghe (Department of Chemistry and Biochemistry, The Ohio State University), Krishna J. Patel (Department of Chemistry and Biochemistry, The Ohio State University), Jane E. Jackman (Department of Chemistry and Biochemistry, The Ohio State University)

Abstract not available online - please check the booklet.

53. Identification of human homologue of mtDNA encoded retrograde signaling molecule, mmu-mito-ncR805

T. L. Mathuram (Department of Biochemistry, University at Buffalo, NY, USA), Y. Perry (Department of Surgery, University at Buffalo, NY, USA), A. Blumental-Perry (Department of Biochemistry, University at Buffalo, NY, USA)

Abstract:
We previously discovered mtDNA-encoded small ncRNA, mmu-mito-ncR805 (1), which increases in Alveolar Epithelial Type II (AETII) during the stress of smoking (SS). Recovery from SS characterizes by distribution of mmu-mito-ncR805 to the nucleus. Redistribution helps to preserve mitochondrial bioenergetics. Mmu-mito-ncR-805 maps to the D-loop of the mtDNA at the beginning of light strand promoter (LSP). Its sequence is not conserved between mouse and human. Nevertheless, identification of human homologue is important because mitochondrial stress and malfunction drives multiple human pathologies. The goal of this study was to identify human homologue of mmu-mito-ncR805 based on genomic location, kinetics and function during stress. Existing databases that contained transcripts originating from the D-loop of human mitochondrial genome were re-analyzed and 4 potential transcripts were identified. Their presence in the relevant human cell lines was confirmed via Northern blot analysis. RNA probes were designed to exclude cross-reactivity to Nuclear Mitochondrial DNA (NUMTS). Mitochondrial origin of the transcripts was further confirmed by their absence in cells with inhibited mitochondrial transcription (2). Mitochondria specific ~260 bp transcript (hsa-mito-ncR-D-loop-805) that originate from the transcription initiation site of human D-loop was further analyzed, and found to increase in its levels during SS. hsa-mito-ncR-D-loop-805 demonstrated dotted pattern throughout the cell, with dots juxtaposed, but not overlapping with mitochondrial protein Tom20. The dots became smaller, and some nuclear localization was detectable during recovery from SS. We therefore identified hsa-mito-ncR-D-loop-805 as a candidate to be a human homologue of mmu-mito-ncR805 retrograde signaling molecule. Our current research is validating hsa-mito-ncR-D-loop-805 levels, kinetics and retrograde signaling function in primary human AETII cells.

References:
1. Blumental-Perry A, Jobava R, Bederman I, Degar AJ, Kenche H, Guan BJ, Pandit K, Perry NA, Molyneaux ND, Wu J, Prendergas E, Ye ZW, Zhang J, Nelson CE, Ahangari F, Krokowski D, Guttentag SH, Linden PA, Townsend DM, Miron A, Kang MJ, Kaminski N, Perry Y, Hatzoglou M. Retrograde signaling by a mtDNA-encoded non-coding RNA preserves mitochondrial bioenergetics. Commun Biol. 2020 Oct 30;3(1):626. doi: 10.1038/s42003-020-01322-4. PMID: 33127975; PMCID: PMC7603330.
2. Gear AR. Rhodamine 6G. A potent inhibitor of mitochondrial oxidative phosphorylation. J Biol Chem. 1974 Jun 10;249(11):3628-37. PMID: 4275428.

Keywords: mmu-mito-ncR805, mtDNA-encoded small ncRNA, retrograde signaling molecule

54. Highly specific, multiplexed isothermal pathogen detection with fluorescent aptamer readout

Lauren M. Aufdembrink (Genetics, Cell Biology and Development, University of Minnesota), Pavana Khan (Genetics, Cell Biology and Development, University of Minnesota), Nathaniel J Gaut (Genetics, Cell Biology and Development, University of Minnesota), Katarzyna P Adamala (Genetics, Cell Biology and Development, University of Minnesota), Aaron E Engelhart (Genetics, Cell Biology and Development, University of Minnesota)

Abstract:
The SARS-CoV-2 pandemic has highlighted the importance of early, rapid and widespread pathogen detection tests that are easily accessible and available to all. We have developed Apta-NASBA, an isothermal, cell-free, synthetic biology-based detection system that combines Nucleic Acid Sequence Based Amplification (NASBA) with transcription of fluorescent aptamers. Despite using the highly active and promiscuous T7 polymerase in the system, we show that the inclusion of a DNA duplex lacking a promoter and unassociated with the amplicon, fully suppresses false positives, enabling a suite of fluorescent aptamers to be used as NASBA tags. With a limit of detection of 1 pM for the E. coli AggR pathogenic template, Apta-NASBA is highly sensitive and can yield multiplexed, multicolor, real-time fluorescent readout. Apta-NASBA can be performed using a variety of equipment, for example a fluorescence microplate reader, a qPCR instrument, or an ultra-low-cost Raspberry Pi-based 3D-printed detection platform employing a cell phone camera module. This method is compatible with field detection and has potential for use in point-of-care SARS-CoV-2 diagnostics. We will discuss our work with Apta-NASBA and other isothermal amplification technologies.

References:
Aufdembrink LM, Khan P, Gaut NJ, Adamala KP, Engelhart AE. Highly specific, multiplexed isothermal pathogen detection with fluorescent aptamer readout. RNA. 2020 Sep;26(9):1283-1290. doi: 10.1261/rna.075192.120.

Keywords: NASBA, T7 RNA polymerase, Isothermal amplification

55. The democratization of RNA therapeutics

John P. Cooke (Center for RNA Therapeutics, Houston Methodist Research Institute, Houston TX), Daniel L. Kiss (Center for RNA Therapeutics, Houston Methodist Research Institute, Houston TX), Ruli Gao (Center for RNA Therapeutics, Houston Methodist Research Institute, Houston TX), Tulsi Damase (Center for RNA Therapeutics, Houston Methodist Research Institute, Houston TX), Yi Liang, Hongye Li (Center for RNA Therapeutics, Houston Methodist Research Institute, Houston TX), Roman Sukhovershin (Center for RNA Therapeutics, Houston Methodist Research Institute, Houston TX)

Abstract:
The development of mRNA vaccines against COVID-19 brought worldwide attention to the transformative potential of RNA-based therapeutics. Integral to this potential, mRNA therapeutics are not solely the province of big biopharma. RNA therapies are a disruptive technology precisely because small biotech startups and academic researchers can rapidly develop innovative and personalized mRNA constructs. However, most of these small groups lack the key competencies to translate their transformational mRNA therapeutics into the clinic. To facilitate the democratization of mRNA therapeutics, we have built the crucial infrastructure to assist smaller groups as they bring their novel ideas to the clinic. In the Texas Medical Center, our fully integrated hospital-based RNA therapeutics program provides a single-entry point with consultation to ensure the seamless development of RNA therapy candidates into transformative drugs. Our RNA Biology and Bioinformatics faculty assist with construct design; our cGMP-trained personnel and clean rooms support the synthesis, purification, and validation of RNA drugs; plus our Nanomedicine colleagues formulate suitable lipid nanoparticles for local or systemic delivery. We have a Comparative Medicine Program with expertise in GLP preclinical studies; a first-in-human clinical trials unit for Phase 1 and 2a studies; and a large hospital system with a clinical research infrastructure that supports Phase 2 and 3 clinical trials. Furthermore, our industry partner manufactures large batches needed for Phase 2, 3 and/or commercialization. To our knowledge, we are the sole academic center with a fully integrated and operational infrastructure to support both academic groups and startups. We intend to support the translational efforts of small groups of scientists as they attain the near limitless potential of RNA Therapeutics.

Keywords: RNA therapeutics, RNA biology, RNA medicine

56. Study of Trm7 interactions with binding partners Trm732 and Trm734 for 2’-O-methylation of the tRNA anticodon loop in yeast

Kellyn Dolezal (Chemistry & Biochemistry; Northern Kentucky University), Thao Le (Chemistry & Biochemistry; Northern Kentucky University), Holly M. Funk (Chemistry & Biochemistry; Northern Kentucky University), Adrian R. Guy (Chemistry & Biochemistry; Northern Kentucky University), Michael P. Guy (Chemistry & Biochemistry; Northern Kentucky University)

Abstract:
Transfer RNAs undergo post-transcriptional modifications, including many which occur at or near the anticodon loop and are necessary for protein synthesis. In Saccharomyces cerevisiae, Trm7 is required for 2’-O-methylation of tRNA and requires interaction with Trm732 and Trm734 for modifications at positions 32 and 34, respectively. These proteins are widely conserved in eukaryotes, and mutations in the human ortholog of TRM7, FTSJ1, can cause non-syndromic X-linked intellectual disability by decreasing or eliminating methylation activity. Through a genetic assay, we have found Trm7 residues required for interaction with Trm732 or Trm734 and the subsequent methylation of tRNA. To verify that this genetic result is due to impaired interaction of Trm7 variants with Trm732 or Trm734, we are performing immunoprecipitation assays. In addition, we have begun to study interactions of the individual proteins and complexes with tRNA through biochemical methods such as electrophoretic mobility shift assays, to determine the role of each protein in tRNA binding. Through this study of Trm7 variants, we can better understand the interaction of Trm7 with Trm734 and Trm732, which will facilitate the study of conserved homologs in humans.

Keywords: tRNA, modifications, Trm7

57. Utilization of high-pressure NMR for the study of the folded and disordered domains of hnRNP A1

Jeffrey D. Levengood (Department of Chemistry, Case Western Reserve University), Julien Roche (Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University), Blanton S. Tolbert (Department of Chemistry, Case Western Reserve University)

Abstract:
The hnRNP A1 protein is involved in numerous processes of nascent RNA transcripts. These roles include, but are not limited to, translational control, splicing regulation, and mRNA stabilization. The protein performs its functions by binding RNA at a high affinity site before recruiting other proteins to aid in its RNA processing activities.

hnRNP A1 is composed of three domains, two structurally identical RRMs that form the N-terminal protein UP1, and a disordered, glycine rich C-terminal domain (G-CTD). The UP1 domain is responsible for binding the nucleic acid substrates while the G-CTD assembles the multi-protein complexes the protein utilizes to carry out its various functions. It is also a low-complexity domain (LCD) capable of mediating liquid-liquid phase separation.

High-pressure NMR spectroscopy is a powerful technique for examining the biophysical nature of proteins. Application of pressure to a protein increases solvent density and pushes the protein towards a lower molar volume, causing folded proteins to unfold. For disordered domains, pressure has been shown to have just minor effects on NMR parameters.

The utilization of high-pressure NMR spectroscopy for the study of full length hnRNP A1 revealed unique properties for both the structured and disordered domains. It revealed the LCD, at standard conditions, adopts a disordered, but compact conformation that is stabilized by polar and electrostatic interactions. The application of pressure results in a disruption of the intra-protein interactions and the compact nature of the domain.

While pressure normally unfolds proteins, the structured UP1 domain failed to unfold at high pressure. To study this phenomenon, UP1 variants were examined to determine what properties of the protein are responsible for its stability at high pressure. Results have revealed that communication between the two RRM domains is vital for maintenance of the protein’s stability during the application of high-pressure.

References:
Levengood JD, Peterson J, Tolbert BS, Roche J. Thermodynamic stability of hnRNP A1 low complexity domain revealed by high-pressure NMR. Proteins. 2021 Jul;89(7):781-791.

Keywords: RRM, IDP, high-pressure NMR

58. Quantitative modeling of ligand-binding thermodynamics in the cyclic RNA-binding oligomeric protein TRAP

Weicheng Li, Andrew Norris (Department of Chemistry and Biochemistry, The Ohio State University), Katie Lichtenthal, Skyler Kelly (Department of Biological Sciences, SUNY Baffulo), Melody L Holmquist, Vicki Wysocki, (Department of Chemistry and Biochemistry, The Ohio State University), Paul Gollnick (Department of Biological Sciences, SUNY Baffulo), Mark P. Foster (Department of Chemistry and Biochemistry, The Ohio State University)

Abstract not available online - please check the booklet.

59. Deploy RNA G1-G4 dendrimers self-assembled from motifs for drug sheltering with controllable layer-by-layer release

Xin Li (Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy, Dorothy M. Davis Heart and Lung Research Institute, James Comprehensive Cancer Center, The Ohio State University), Peixuan Guo (Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy, Dorothy M. Davis Heart and Lung Research Institute, James Comprehensive Cancer Center, The Ohio State University)

Abstract:
Dendrimers are polymeric molecules with defined and monodisperse branched structures, which have been used as drug delivery platforms due to their unique properties. Taking advantage of the self-assembly and programmability property of RNA, we used three-way junction RNA nanoparticles as the building block for dendrimer construction to achieve simple synthesis procedure and favorable biodistribution profile. Oligo ribonucleotides produced by solid phase chemical synthesis allow the large-scale manufacture of homologous RNA dendrimers. Employing concepts from RNA nanotechnology enabled the controllable production of four generations of RNA dendrimers from G1, G2, G3, to G4 with temperature-triggered layer-by-layer release capability. The conjugation of various functional groups into individual RNA strands and the incorporation of functionalized RNA strands into the dendrimers at different sites have been reported. RNA dendrimers conjugated with multiple copies of anticancer drug, paclitaxel, showed comparable cancer cell inhibition effect to free drugs. Furthermore, the encapsulation of cell binding ligands and hydrophobic drugs within the dendrimer significantly reduced the efficiency of cell binding and protein binding respectively, demonstrating the shielding effect of RNA dendrimers. The results imply a potential application of RNA dendrimer for delivery, shielding and controlled release of hydrophobic drugs in vivo.

References:
1. X. Li, M. Vieweger and P. Guo, Nanoscale, 2020, 16514–16525.
2. S. Guo, M. Vieweger, K. Zhang, H. Yin, H. Wang, X. Li, S. Li, S. Hu, A. Sparreboom, B. M. Evers, Y. Dong, W. Chiu and P. Guo, Nat. Commun., 2020, 11, 972.
3. A. Sharma, F. Haque, F. Pi, L. S. Shlyakhtenko, B. M. Evers and P. Guo, Nanomedicine Nanotechnology, Biol. Med., 2016, 12, 835–844.

Keywords: RNA nanotechnology, RNA dendrimer, Drug delivery and shielding

60. Androgen Receptor messenger RNA Metabolism

Eviania M. Likos (Biological, Geological, and Environmental Sciences, Cleveland State University), Girish C. Shukla (Biological, Geological, and Environmental Sciences, Cleveland State University)

Abstract not available online - please check the booklet.

61. Pum3 as a molecular player in transcriptional termination and R-Loop-Associated DNA Damage

chenyu Lin (The James Comprehensive Cancer Center, The Ohio State University), Wayne Miles (The James Comprehensive Cancer Center, The Ohio State University)

Abstract:
Pum3 as a molecular player in transcriptional termination and R-Loop-Associated DNA Damage

Pum3 was reported to correlate with resistance to genotoxic exposure ssDNA break via preventing PARP1 degradation by caspase3. Given it is vital for the maintenance of homeostasis that DSBs within transcriptionally active regions undergo accurate repair. However, it remains unknown how Pum3 modulates DSBs response and DNA repair. Our research identified Pum3 as RNA/DNA hybrid interactome that promotes R-loop and transcriptional termination. Pum3 is recruited to the DSB site in a DNA-RNA-hybrid-dependent manner, as playing a critical role in promoting Pol III-mediated R-loop processing and imitating subsequent homologous recombination-mediated repair. Pum3-deficient induces defective HR, accumulation of unrepaired DSBs. Pum3 is overexpressed in PRAD and LIHC cancers, and higher expression of Pum3 correlates with lower survival probability in these cancers. Depletion of Pum3 increases the sensitivity of cancer cells to DNA damage-based therapy-cisplatin. Thus, our data indicated that the function of Pum3 in the presence of R-loops around DSBs within transcriptionally active regions promotes accurate repair of DSBs, which may have implications for cancer therapy.

References:
hPuf-A/KIAA0020 modulates PARP-1 cleavage upon genotoxic stress. (PMID: 21266351)

Keywords: Pum3, R-loop , DNA damage

62. Coacervate rescue of nucleic acid enzyme activity damaged by UV

Haiyun Liu (Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA), Philip C Bevilacqua (Department of Chemistry; Center for RNA Molecular Biology; Department of Biochemistry, Microbiology, and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA)

Abstract:
According to the RNA world hypothesis, RNA had the roles of catalyst and genetic information carrier in the emergence of life. However, lack of proofreading in RNA replication and the unstable chemical nature of RNA compared to DNA make it more challenging for RNA to retain its catalytic activity. Additionally, ribozymes were under strong UV radiation of the early earth which lacked an ozone layer. On the other hand, membraneless compartments such as coacervates have been hypothesized as plausible prebiotic compartments due to their ability to enrich cations, nucleotides and RNA. Moreover, enhancements of activity of various ribozymes by coacervates have been described in previous studies. We hypothesize that coacervates can rescue the activity of intact ribozymes, as well as damaged, mutated, and fragmented ones. To test this hypothesis, we are using UV light to damage a DNAzyme, and attempting rescuing it with PDAC/D10 coacervate. Our latest design and results will be presented.

References:
Poudyal, R.R., Guth-Metzler, R.M., Veenis, A.J. et al. Template-directed RNA polymerization and enhanced ribozyme catalysis inside membraneless compartments formed by coacervates. Nat Commun 10, 490 (2019). https://doi.org/10.1038/s41467-019-08353-4

Keywords: Astrobiology, Nucleic acid enzyme, Coacervate

63. Investigation of structural switch that regulates pre-miR-92a processing by NMR

Yaping Liu (University of Michigan), Sarah Keane (University of Michigan)

Abstract:
MicroRNAs (miRNAs) regulate gene expression in a variety of biological pathways such as development and tumorigenesis. miRNAs are initially expressed as a long primary transcripts (pri-miRNA) which are processed to yield pre-miRNAs and ultimately mature miRNAs. The miR-17-92a cluster, also known as ‘oncomiR-1’ is a polycistronic pri-miRNA that plays a pivotal regulatory role in cellular processes, including the cell cycle, proliferation and apoptosis. OncomiR-1 includes six constitute miRNAs, each processed with different efficiencies as a function of both developmental time and tissue type. However, the structural mechanism that regulate the differential processing still remain unclear.
NMR is a key technique that has significantly advanced our understanding of RNA structure and dynamics. In vitro processing assays indicate that pre-miR-92a processing is inhibited relative to other miRs within oncomiR-1. We are interested in uncovering the structural basis for the differential processing by NMR. NPSL2 has been identified as an inhibitor of pre-miR-92a processing. Therefore, in this work we determined the solution structure of NPSL2. Our data suggest that NPSL2 is dynamic, and we hypothesize that the dynamics are linked to regulation of processing. Our ongoing efforts will investigate the mechanism of the structural switch that promotes premiR-92a processing in the context of the full-length oncomiR-1 RNA.

References:
1. Kim, V. N., Han, J. & Siomi, M. C. Nat Rev Mol Cell Biol (2009) 10, 126-139. doi:10.1038/nrm2632.
2. Mogilyansky, E. & Rigoutsos, I. Cell Death Differ (2013) 20, 1603-1614. doi:10.1038/cdd.2013.125. PMC3824591.
3. He, L. et al. Nature (2005) 435, 828-833. doi:10.1038/nature03552. PMC4599349.
4. O'Donnell, K. A., Wentzel, E. A., Zeller, K. I., Dang, C. V. & Mendell, J. T. Nature (2005) 435, 839-843. doi:10.1038/nature03677.
5. Chakraborty, S. & Krishnan, Y. Nucleic Acids Res (2017) 45, 9694-9705. doi:10.1093/nar/gkx613. PMC5766152.
6. Chaulk, S. G., Xu, Z., Glover, M. J. & Fahlman, R. P.. Nucleic Acids Res (2014) 42, 5234-5244. doi:10.1093/nar/gku133. PMC4005684.

Keywords: MicroRNNAs, oncomiR-1, NPSL2

64. Selectivity mechanisms of an aminoacyl-tRNA trans-editing factor

Xiao Ma (Department of Chemistry and Biochemistry, the Ohio State University), Marina Bakhtina (Department of Chemistry and Biochemistry, the Ohio State University), Karin Musier-Forsyth (Department of Chemistry and Biochemistry, the Ohio State University), Mark P. Foster (Department of Chemistry and Biochemistry, the Ohio State University)

Abstract:
Aminoacyl-tRNA synthetases (aaRSs) are responsible for charging their cognate tRNAs with the correct or cognate amino acid. The structural features of aaRS catalytic domains provide a high degree of selectivity for both the amino acid and tRNA; however, given the similar stereochemical properties of many amino acids, errors in tRNA charging can occur, especially for the smaller and isometric amino acids. Organisms in all three domains of life possess editing enzymes that function in trans to deacylate mischarged tRNAs. Caulobacter crescentus ProXp-ala recognizes the terminal base pair, C1:G72, on the acceptor stem of tRNAPro to deacylate mischarged Ala-tRNAPro but not Ala-tRNAAla. We used NMR spectroscopy, binding and activity assays to examine the structural details of this discrimination. We also found that the A76 of tRNAPro was essential for its binding to ProXp-ala, thus supporting a previously proposed interaction with a highly conserved Lys. Ongoing work continues to refine a 3D model of the enzyme-product complex of ProXp-ala bound to a tRNAPro acceptor stem. These studies will improve our understanding of substrate recognition by a bacterial trans-editing factor with the potential to inform new antibiotic drug design.

References:
Das M., Vargas-Rodriguez O., Goto Y., Novoa, E., Pouplana L., Suga H., Musier-Forsyth K., (2014). Distinct tRNA recognition strategies used by a homologous family of editing domains prevent mistranslation. Nucl. Acids Res. 42 (6):3943-3953.

2. Ling J, Reynolds N, Ibba M (2009) Aminoacyl-tRNA synthesis and translational quality control. Annu Rev Microbiol 63:61–78.

3. Danhart, E. M., Bakhtina, M., Cantara, W. A., Kuzmishin, A. B., Ma, X., Sanford, B. L., Košutić, M., Goto, Y., Suga, H., Nakanishi, K., et al. (2017) Conformational and chemical selection by a trans - acting editing domain. Proc. Natl. Acad. Sci. 114, 6774–6783.

Keywords: NMR, Protein-RNA

65. Human spliceosomal snRNA sequence variants generate variant spliceosomes

Justin W. Mabin (Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health), Peter W. Lewis (Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health), David A. Brow (Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health), Heidi Dvinge (Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health)

Abstract:
Human pre-mRNA splicing is primarily catalyzed by the major spliceosome, comprising five small nuclear ribonucleoprotein complexes, U1, U2, U4, U5, and U6 snRNPs, each of which contains the corresponding U-rich snRNA. These snRNAs are encoded by large gene families exhibiting significant sequence variation, but it remains unknown if most human snRNA genes are untranscribed pseudogenes or produce variant snRNAs with the potential to differentially influence splicing. Since gene duplication and variation are powerful mechanisms of evolutionary adaptation, we sought to address this knowledge gap by systematically profiling human U1, U2, U4 and U5 snRNA variant gene transcripts. We identified 55 transcripts that are detectably expressed in human cells, 38 of which incorporate into snRNPs and spliceosomes in 293T cells. All U1 snRNA variants are more than one thousand-fold less abundant in spliceosomes than the canonical U1, whereas at least 1% of spliceosomes contain a variant of U2 or U4. In contrast, eight U5 snRNA sequence variants occupy spliceosomes at levels of 1 to 46%. Furthermore, snRNA variants display distinct expression patterns across five human cell lines and adult and fetal tissues. Different RNA degradation rates contribute to the diverse steady state levels of snRNA variants. Our findings suggest that variant spliceosomes containing non-canonical snRNAs may contribute to different tissue- and cell-type specific alternative splicing patterns.

Keywords: snRNA, snRNA biogenesis, snRNA variants, spliceosomes, pre-mRNA splicing

66. Deducing the role of ADR-2 in immune gene regulation in C. elegans

Ananya Mahapatra (Genome, Cellular and Developmental Biology, Indiana University), Sarah Deffit (Medical Sciences Program, Indiana University School of Medicine), Pranathi Vadlamani (Medical Sciences Program, Indiana University School of Medicine), Heather A. Hundley (Medical Sciences Program, Indiana University School of Medicine),

Abstract not available online - please check the booklet.

67. Human patient derived iPSCs as a system for studying cardiac dysfunctions in Myotonic Dystrophy type 1

Sarah N. Matatov (Department of Biochemistry University of Illinois, Urbana-Champaign, Illinois, USA), Chaitali Misra (Department of Biochemistry University of Illinois, Urbana-Champaign, Illinois, USA), Auinash Kalsotra (Department of Biochemistry, Cancer CenterIllinois, Carl R. Woese Institute of Genomic Biology, University of Illinois, Urbana-Champaign, Illinois, USA )

Abstract:
Myotonic Dystrophy type 1 (DM1), is the most prevalent form of adult-onset muscular dystrophy. DM1 is caused by a CTG trinucleotide repeat expansion in the 3’-UTR of the DMPK gene. Over 80% of DM1 patients experience heart dysfunctions making this the second leading cause of death for affected individuals. The CUG expanded repeat in RNA creates a toxic gain-of-function phenotype, which causes RNA aggregation into ribonuclear foci. These RNA foci sequester Muscleblind-like Splicing Regulators (MBNL) and cause over-expression of the CUGBP Elav-Like Family Member 1 (CELF1) family of splicing factors. Our lab recently demonstrated that aberrant expression of a non-muscle splice isoform of RNA-binding protein RBFOX2 triggers the characteristic cardiac conduction delay, atrioventricular heart blocks, and spontaneous arrhythmogenesis in DM1 hearts. To further study the mechanisms by which non-muscle RBFOX2 alters the electrophysiological properties and thereby induces cardiac arrhythmias in DM1, we generated a human cardiomyocyte cell culture model for DM1. We acquired induced pluripotent stem cells (iPSC) from a healthy donor, a patient with a mild case of DM1 (238 repeats), and another patient with severe case of DM1 (1150 repeats). We successfully established a robust protocol for differentiating these iPSC lines into beating cardiomyocytes with reliable morphologic, molecular, and functional features. Importantly, we demonstrate that the differentiated experimental cells faithfully recapitulate the classical characteristics of DM1, including ribonuclear foci formation, MBNL sequestration, misregulation of alternative splicing, cardiac conduction delay, and contractility defects. Using these human patient-derived cardiac culture systems, we are now investigating the mechanistic and functional roles of specific RNA binding proteins in DM1 cardiac pathogenesis.

Keywords: Muscular Dystrophy, Cardiac Dysfunction, iPSCs

68. Title not available online - please see the booklet.

Chaitali Misra (Departments of Biochemistry, University of Illinois, Urbana-Champaign), Ullas V. Chembazhi (Departments of Biochemistry, University of Illinois, Urbana-Champaign), Sarah N. Matatov (Departments of Biochemistry, University of Illinois, Urbana-Champaign), Auinash Kalsotra (Departments of Biochemistry, Cancer CenterIllinois, Carl R. Woese Institute of Genomic Biology, University of Illinois, Urbana-Champaign)

Abstract not available online - please check the booklet.

69. Calculations of theoretical pKa values for modified nucleobases

Alan J. Mlotkowski (Chemistry Department - Wayne State University ), Evan Jones (Chemistry Department - Wayne State University ), H. Bernhard Schlegel (Chemistry Department - Wayne State University ), Christine S. Chow (Chemistry Department - Wayne State University )

Abstract not available online - please check the booklet.

70. GSK3α plays a vital role in male fertility by regulating m6A levels in testis

Rushdhi Rauff (Department of Chemistry and Biochemistry, Kent State University), Souvik Dey (Department of Biological Sciences, Kent State University), Wesam Nofal (Department of Biological Sciences, Kent State University), Mustfa Kabi (Department of Biological Sciences, Kent State University), Srinivasan Vijayaraghavan (Department of Biological Sciences, Kent State University), Sanjaya Abeysirigunawardena (Department of Chemistry and Biochemistry, Kent State University)

Abstract:
N6-methyladenosine (m6A) is the most abundant nucleotide modification observed in eukaryotic mRNA. N6-methylation levels are dynamic in mRNA, where methylations are added and removed by proteins that are known as writers and erasers, respectively. FTO (Fat mass and obesity-associated protein) is one of the most well-studied methyl eraser proteins found in eukaryotes. It has been known that in stem cells, FTO is a substrate of GSK3 protein. GSK3 enzyme, a serine/threonine protein kinase, is ubiquitous and is expressed as two paralogs, GSK3α and GSK3β, encoded by two genes. The only phenotype of global deletion of Gsk3α is male infertility. Here we hypothesize that GSK3α plays a similar role in controlling m6A levels in testis by interacting with FTO. Our experiments show that FTO is localized to post-meiotic spermatogenic cells within the seminiferous tubules. There is no visible staining for FTO in Sertoli cells, spermatogonia, myoid, and Leydig cells in the interstitial spaces of the seminiferous tubules. Co-immunoprecipitation of FTO with GSK3α, but not with GSK3β was observed when immunoprecipitation was carried out for GSK3α and GSK3β followed by western blots using FTO primary antibodies. A marked increase in FTO expression was seen in day 20 testis up to day 40 adult testis; an expression pattern characteristic of proteins expressed in post-meiotic developing spermatocytes and spermatids. Using HPLC and ELISA-based methods, we observed that m6A levels are significantly lower in GSK3α KO mice compared to WT mice. Hence, our experimental findings illustrate that GSK3α plays a main role in male fertility by regulating m6A levels in testis.

Keywords: m6A, nucleotide modifications, epitranscriptomics

71. Elucidating the mechanisms of regulation for MDM2 alternative splicing

Matias Montes (Center for Childhood Cancer and Blood diseases Nationwide Childrens Hospital), Dawn Chandler (Center for Childhood Cancer and Blood diseases Nationwide Childrens Hospital)

Abstract:
MDM2, a well-known negative regulator of the tumor suppressor protein p53 undergoes complex alternative splicing. One of its spliced isoforms, MDM2-ALT1 comprised of exons 1 through 3 and 12, is highly expressed in several cancers such as lung carcinoma, liposarcoma and rhabdomyosarcoma (RMS). In RMS, MDM2-ALT1 expression correlates with a poor disease prognosis. MDM2-ALT1 expression is also upregulated under conditions of cellular genotoxic stress. The way that MDM2 splicing is controlled is not well understood. Here we explore the possibility that a nuclear miRNA, miR29b, interplays with RNA binding proteins to regulate MDM2 splicing.

miR-29b is part of the miR-29 family, and has been reported as one of the miRNAs that localize to the nuclear compartment of cells. However, its function there is still unknown. Additionally, miR-29b has been shown to act as a tumor suppressor. In RMS, we have shown how miR-29b levels are downregulated in both cell lines as well as in patient samples. Here, we show how miR-29b is downregulated after genotoxic stress, which inversely correlates with MDM2-ALT1 expression, similarly to RMS. Furthermore, we show how miR-29b influences the alternative splicing of MDM2, which is not through canonical gene-silencing of splicing factors. We have identified predicted binding sites for the miR-29b in the regulated region of MDM2 pre-mRNA. Moreover, we demonstrate that miR-29b directly binds to the MDM2 pre-mRNA and that the site present in the exon 12 is important for alternative splicing regulation. Finally, we show how Argonaute 1 antagonizes miR-29b influence over the MDM2 alternative splicing, and interaction between AGO1 and miR-29b is increased after genotoxic stress. In summary, in this work we report for the very first time how a nuclear miRNA has direct involvement regulating the alternative splicing of a gene.

Keywords: alternative splicing, miRNAs, MDM2

72. Identifying novel m6A binding proteins using a peptide as a bait which was enriched in a phage display experiment.

David Morgan (Department of Chemistry and Biochemistry - Kent State University ), Rushdhi Rauff (Department of Chemistry and Biochemistry - Kent State University ), Sanjaya Abeysirigunawardena (Department of Chemistry and Biochemistry - Kent State University )

Abstract:
N6-methyladenosine (m6A) is the most abundant nucleotide modification in eukaryotic mRNA. The m6A modification can be added, removed, and recognized by m6A writers, erasers, and readers, respectively. Due to the presence of many m6A modifications in some mRNAs, likely there are more m6A binding proteins that can interact site-specifically. Protein pull-down assays were performed using various methylated and unmethylated RNA targets to identify proteins that are bound to methylated RNA with sequence and structural specificity. Lysates from various cancer cell-lines were used for these pull-down assays. Interestingly, the enriched proteins have sequence similarity to dodecamer peptides selected from phage display experiments using same RNA baits. Our experiments show that m6A readers interacts with RNA using unique strategies.

Keywords: m6A, Nucleotide modifications, Epitranscriptomics

73. Long noncoding RNAs are abundant targets of ADAR3 in glioblastoma

Priyanka Mukherjee (Cell, Molecular and Cancer Biology, Indiana University School of Medicine), Heather A. Hundley (Medical Sciences Program, Indiana University School of Medicine)

Abstract not available online - please check the booklet.

74. Towards structural characterization of RNA-only complexes via small-angle scattering techniques

Aldrex Munsayac (Department of Chemistry, University of Michigan), Ian Hall (Department of Chemistry, University of Michigan), Sarah C. Keane (Department of Chemistry and Program in Biophysics, University of Michigan)

Abstract:
Noncoding RNAs constitute the majority of the transcribed human genome and play a multitude of roles in biological processes; having been shown to regulate gene expression at the levels of transcription, RNA processing, and translation.1 In the foodborne pathogen Listeria monocytogenes, two riboregulators—the prfA RNA thermosensor (RNAT) and the SreA riboswitch—work in tandem to regulate the expression of PrfA, the major regulator of Listeria virulence gene expression.2 However, the precise molecular mechanisms governing the interaction between the prfA RNAT and SreA riboswitch remains undefined. In order to understand how these two cis-regulatory RNA elements are able to function in trans, elucidation of their structure at both the global and local scale is necessary. RNA-only structures account for only 1% of the three-dimensional structures deposited in the protein data bank.3 Furthermore, we expect that both the prfA RNAT and the SreA riboswitch undergo significant remodeling upon interaction, necessitating the development and implementation of new approaches for determining the structures of RNA-RNA complexes. To address this, we propose to employ a hybrid structural biology approach using small-angle X-ray/neutron scattering (SAXS/SANS) techniques complemented with NMR spectroscopy to unambiguously probe RNA-RNA interactions. Here, we lay the groundwork for the development of contrast variation SANS (CV-SANS) for RNA-only complexes. Using the HIV-1 RNA dimerization initiation site (DIS) kissing complex as a model system, we have collected SAXS data and successfully reconstructed the molecular envelope of the complex, which will serve as the foundation for our initial CV-SANS studies. CV-SANS coupled with NMR spectroscopy has the potential to solve very large, dynamic RNA structures.

References:
1. Cech, T.R.; Steitz, J.A. The noncoding RNA revolution—trashing old rules to forge new ones. Cell. 2014, 35, 77-94.

2. Loh, E.; Dussurget, O.; Gripenland, J;, Vaitkevicius, K.; Tiensuu, T.; Mandin, P.; Repoila, F.; Buchrieser, C.; Cossart, P.; Johansson, J. A trans-acting riboswitch controls expression of the virulence regulator PrfA in Listeria monocytogenes. Cell. 2009, 139(4), 770-779.

3. Das, R. RNA structure: a renaissance begins?. Nature Methods. 2021, 18(5), 439-439.

Keywords: Small-angle scattering, RNA-RNA interactions, RNA structural biology

75. Proximity labeling to monitor in vivo protein dynamics during nonsense-mediated mRNA decay

Sarah L.H. Nock (Department of Genetics and Genome Sciences, Case Western Reserve University), DaJuan L. Whiteside (Department of Genetics and Genome Sciences, Case Western Reserve University), Kristian E. Baker (Department of Genetics and Genome Sciences, Case Western Reserve University)

Abstract not available online - please check the booklet.

76. Title not available online - please see the booklet.

Elizabeth A. Oser (Department of Chemistry and Biochemistry, The Ohio State University), Tyler McClain (Department of Chemistry and Biochemistry, The Ohio State University), Jane E. Jackman (Department of Chemistry and Biochemistry, The Ohio State University)

Abstract not available online - please check the booklet.

79. A key to understanding RNA’s interactome: photocleavable probes for spatiotemporal control of RNA

Marta Rachwalak (Chemistry, Carnegie Mellon University), Avleen K. Chawla (Chemistry, Carnegie Mellon University), Megan L. Van Horn (Chemistry, Carnegie Mellon University), Anna M. Kietrys (Chemistry, Carnegie Mellon University)

Abstract:
Caging is one of the key methods for imaging and studying interactions and structural aspects of RNA inside the cell. Despite the rapid improvements in caging and cloaking techniques, scientists still struggle to develop universal and effective tools for understanding the complexity and dynamics of the RNA interactome in cellulo. To overcome this absence of appropriate tools, we designed and synthesized a set of photoreversible probes for studying RNA’s function and interactions within cells. An introduction of photocleavable protecting groups (PPGs) ensures control over the biological function of various RNAs (e.g., siRNA, mRNA) and allows for structure and function reactivation, in a spatiotemporal manner, in the living cell. Using the same set of probes in living cells may help to map short- and long-distance interactions in RNA complexes.
Herein, we present a synthesis of photocleavable (INPhRNA) probes designed to block RNA’s biological function through acylation of 2′-OH groups in single stranded regions. Protection of 2’-OH moieties significantly disrupts RNA structure and as such its function. The RNA function may be fully restored via UV-irradiation without any cellular damage. The photosensitive residue of the probes consists of PPG units connected by linkers. By modulating the type of linker and its length, we are able to affect both the solubility and reactivity of the probe. To show the biological applicability of the proposed strategy, we tested the probe’s cytotoxicity and potential for spatiotemporal control over siRNA and mRNA. We believe that the developed caging probes may serve as an efficient method for postsynthetic modification of RNA to protect them from degradation and direct their cellular activity. Additionally, they may be used as tools to explore RNA interactome complexity in vivo.

Keywords: RNA, photocleavable protecting group, RNA interactome

80. Transcriptome-wide analysis of ADAR3 binding and regulation of ADAR1 editing activity in glioblastoma cells

Reshma Raghava Kurup (Genome, Cell and Developmental Biology Program, Indiana University, Bloomington ), Emilie Oakes (Genome, Cell and Developmental Biology Program, Indiana University, Bloomington ), Aidan Manning (Medical Sciences Program, Indiana University School of Medicine, Bloomington IN), Pranathi Vadlamani (Medical Sciences Program, Indiana University School of Medicine, Bloomington IN), Heather A. Hundley (Medical Sciences Program, Indiana University School of Medicine, Bloomington IN)

Abstract not available online - please check the booklet.

81. Pumilio regulates Dicer1 protein and miRNA levels in human cells

Swetha Rajasekaran (Department of Molecular Genetics, The Ohio State University), Wayne Miles (Department of Cancer Biology and Genetics, The Ohio State University)

Abstract:
DICER1 is an endoribonuclease involved in processing of mature miRNAs. Maintaining homeostasis of DICER1 protein levels has shown to be important as changes in Dicer1 levels can affect cellular phenotypes, and drive tumor growth. Mutations within the DICER1 gene cause Dicer1 syndrome – an inherited disorder that results in the development of uncommon cancerous and benign tumors. Most DICER1 mutations occur in the coding sequence of the transcript, however around 30% of the Dicer1 syndrome patients do not have a mapped mutation within this region. By analyzing sequences from a large cohort of Dicer1 syndrome patients, our lab has identified an A-to-G mutation in the 3’ untranslated region (3’ UTR) of DICER1. Motif mapping studies have shown that this mutation disrupts the binding site of the Pumilio (PUM) RNA-binding protein (RBP). The PUM family of proteins bind a consensus PUM Regulatory Element (PRE) within the 3’UTR of the mRNA to affect levels of protein translation and mRNA turnover of the substrate. PUM proteins are generally considered to be translational repressors that bind their targets and reduce the translation efficiency and ultimately the protein expression of the transcript. Surprisingly, on depleting PUM proteins using shRNA or CRISPR, we observed a decrease in DICER1 protein levels compared to WT, contrary to the expected model of PUM-mediated translational repression. Luciferase assays with WT and PRE-mutant DICER1 3’UTR fragments showed that this regulation is PRE-mediated and caused by direct binding. To test the importance of PUM-mediated regulation of Dicer1, we made knock-in of PRE-mutations in the endogenous DICER1 3’UTR in human cell lines using CRISPR. We then measured how this mutation affected growth rate, invasion and migration and compared with WT cells. These experiments showed that an isolated mutation in the DICER1-PRE resulted in slower growth, lesser invasion and decreased rate of migration in these cells. DICER1 expression is a double-edged sword in cancer and it is important for the cells to maintain the right amount of DICER1 making it hard to develop therapeutic strategies directly targeting DICER1. Understanding PUM-mediated post-transcriptional regulation of DICER1 may provide opportunities to target DICER1 activity in tumors.

Keywords: Pumilio, DICER1

82. A Zinc-Responsive Ribosomal Protein Regulation Mechanism in E. coli

Rebecca A. Rasmussen (Interdisciplinary Biological Sciences Program, Northwestern Univeristy), Suning Wang (Department of Chemistry, Northwestern University), Julius B. Lucks (Department of Chemical and Biological Engineering, Northwestern University), Thomas V. OHalloran (Department of of Microbiology & Molecular Genetics, Michigan State University)

Abstract not available online - please check the booklet.

83. Impact of RNA structure on histone lysine specific demethylase 1 (LSD1) activity and nucleosome engagement.

Dulmi Senanayaka (Dept of Chemistry, Marquette University), William J Martin (Dept of Biochemistry, Vanderbilt University), Danyun Zeng (Dept of Chemistry, Marquette University), Manuel Ascano Jr. (Dept of Biochemistry, Vanderbilt University), Nick Reiter (Dept of Chemistry, Marquette University)

Abstract not available online - please check the booklet.

84. Characterization of lipoprotein-bound microRNAs

Guido Rossi-Herring (Bellaterra Campus, Universitat Autnoma de Barcelona, Cerdanyola del Valls, 08041 Barcelona, Spain), David de Gonzalo-Calvo (Institute of Biochemical Research of Barcelona IIBB-Spanish National Research Council CSIC, Barcelona, Spain; Lipids and Cardiovascular Pathology Group, Biomedical Research Institute Sant Pau IIB), Jose Luis Snchez-Quesada (Cardiovascular Biochemistry Group, Research Institute of the Hospital de la Santa Creu i Sant Pau IIB Sant Pau, 08041 Barcelona, Spain)

Abstract:
MicroRNAs (miRNAs) are small non-coding RNAs that control gene expression at posttranscriptional level through targeting of messenger RNA (mRNA) and play an important role in physiological and pathological processes such as cancer, cardiovascular disease and other metabolic diseases. miRNAs have been described in extracellular space and blood transported by extracellular vesicles and proteins. Extracellular miRNAs are also carried by lipoproteins, HDL and LDL; however, technical issues limited their evaluation. Here, we describe the optimal methodology to analyze miRNAs expression in lipoprotein fractions. Additionally, we explored the HDL, LDL and VLDL-miRNA signatures in pure fractions of each lipoprotein. Lipoproteins were isolated from serum by ultracentrifugation and subsequent purification using fast protein liquid chromatography (FPLC). First, the amount of protein needed for RNA isolation in lipoproteins was determined from three serum pools (n=3). 300 μg of protein resulted in the optimal amount of protein for miRNAs expression profiling in lipoproteins. Second, a total of 179 miRNAs were evaluated in the three lipoprotein fractions isolated from serum pools (n=6). A high correlation was observed between VLDL and HDL-miRNA signatures. The LDL-miRNA signature was clustered in a distinctive group. VLDL and HDL shared loaded miRNAs, as well as carried specific miRNAs. Otherwise, LDL did not transport specific miRNAs. Three miRNAs resulted to be highly-expressed in the three lipoproteins (miR-125a-5p, miR-1260a and miR-335-3p). Pathway analysis showed common molecular pathways regulated by the miRNAs expressed in the lipoprotein fractions. Two sequence motifs were detected in HDL-miRNAs, suggesting a possible sorting mechanism for miRNAs. Collectively, these findings support the idea that lipoproteins are carriers of circulating miRNAs. Of note, we show for the first time the miRNA signature of the VLDL fraction. In addition, we describe a new standardization method for miRNA detection in lipoproteins.

References:
Valadi, H. et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007).
Vickers, K. C., Palmisano, B. T., Shoucri, B. M., Shamburek, R. D. & Remaley, A. T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 13, 423–433 (2011).
Wagner, J. et al. Characterization of Levels and Cellular Transfer of Circulating Lipoprotein-Bound MicroRNAs. Arterioscler. Thromb. Vasc. Biol. 33, 1392–1400 (2013).
Gonzalo-Calvo, D. et al. Translating the microRNA signature of microvesicles derived from human coronary artery smooth muscle cells in patients with familial hypercholesterolemia and coronary artery disease. J. Mol. Cell. Cardiol. 106, 55–67 (2017).

Keywords: miRNAs, lipoprotein, signature

85. Investigating how upstream open reading frame length regulates re-initiation and mRNA stability

Paul J. Russell (Cellular, Molecular, and Biochemical Sciences Program, Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University, Columbus, OH 43210), Elaina P. Boyle (Cellular, Molecular, and Biochemical Sciences Program, Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University, Columbus, OH 43210), Michael G. Kearse (Cellular, Molecular, and Biochemical Sciences Program, Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University, Columbus, OH 43210)

Abstract:
Approximately 50% of all mammalian transcripts contain at least one upstream open reading frames (uORFs). Since uORFs exist in the 5' untranslated region (UTR), they are classically thought to inhibit translation of the downstream primary open reading frame (ORF) as they directly compete for the scanning ribosome. However, it is well accepted that ribosomes can re-initiate after translating uORFs. Additionally, uORFs have been implicated in regulating the stability of the mRNA via nonsense-mediated mRNA decay (NMD) and re-initiation may be a method used to evade NMD. Here we use re-initiation specific luciferase reporters with custom uORF sequences of different lengths in rabbit reticulocyte lysate and in HeLa cells to assess uORF length dependency on re-initiation and mRNA stability. Our data shows our custom uORFs capture all detectable scanning ribosomes and that re-initiation is more efficient after translating short uORFs. Using class I-IV viral internal ribosome entry site (IRES) elements to drive initiation with varying smaller sets of initiation factors, our data also supports the model that stubbornly-bound initiation factors aid re-initiation. We are now primed to test if re-initiation is a robust method for mRNAs to use to evade NMD. Together, this study will expand our understanding of how uORFs influence translational control and mRNA stability in mammalian cells.

Keywords: translational control, eIF, ribosome

86. PYM controls Exon Junction Complex occupancy at non-canonical positions

Manu Sanjeev (Molecular Genetics, The Ohio State University), Lauren Woodward (Molecular Genetics, The Ohio State University), Robert Patton (Physics, The Ohio State University), Ralf Bundschuh (Physics, The Ohio State University), Guramrit Singh (Physics, The Ohio State University)

Abstract:
As mRNAs undergo the splicing reaction in the nucleus, the spliceosome deposits the Exon Junction Complex (EJC) ~24 nucleotides upstream of mRNA exon-exon junctions. EJCs thus deposited remains stably bound to mRNA and modulates mRNA fate at multiple post-transcriptional steps until its disassembly during translation and is essential for viability in vertebrates[1]. A ribosome-associated factor called PYM, which directly binds to the Y14/MAGOH heterodimer of the EJC core, is reported to aid in EJC disassembly during translation[2]. However, such a role for PYM is contradicted by the observation that Drosophila PYM null mutants are fully viable[3]. Furthermore, PYM levels are sub-stoichiometric to both EJC and ribosomes. Here, we have investigated the role of PYM in human cells by using a previously reported MAGOH mutant that assembles into EJC but is impaired in PYM interaction. Using RNA immuno-precipitation in tandem and high-throughput sequencing (RIPiT-Seq) to identify footprints of EJC from human embryonic kidney (HEK293) cells, we find that EJCs lacking PYM interaction show no defect in translation dependent disassembly. We find that the loss of EJC-PYM interaction leads to increased EJC footprints away from the -24 position (non-canonical EJC binding sites). Surprisingly, PYM interaction deficient EJC is also enriched on unspliced transcripts from many single-exon genes that do not normally contain the EJC. Our findings suggest a model for PYM function where PYM prevents spontaneous, splicing-independent deposition of EJC at non-canonical sites across the transcriptome. We are currently testing this model using biochemical and transcriptomic analyses. Our work reveals a potential mechanism for EJC binding at non-canonical sites and has important implications for viral pathogenesis as PYM is targeted by Kaposi Sarcoma-associated Herpes Virus to enhance viral RNA translation and by flaviviruses to promote viral replication via NMD inhibition[4].

References:
1] L. A. Woodward, J. W. Mabin, P. Gangras, and G. Singh, “The exon junction complex: a lifelong guardian of mRNA fate: EJC: assembly, structure, and function,” WIREs RNA, vol. 8, no. 3, p. e1411.
[2] N. H. Gehring, S. Lamprinaki, A. E. Kulozik, and M. W. Hentze, “Disassembly of Exon Junction Complexes by PYM,” Cell, vol. 137, no. 3, pp. 536–548, May 2009.
[3] S. Ghosh, A. Obrdlik, V. Marchand, and A. Ephrussi, “The EJC Binding and Dissociating Activity of PYM Is Regulated in Drosophila,” PLoS Genet, vol. 10, no. 6, p. e1004455, Jun. 2014.
[4] M. Li et al., “Identification of antiviral roles for the exon–junction complex and nonsense-mediated decay in flaviviral infection,” Nat Microbiol, vol. 4, no. 6, pp. 985–995, Jun. 2019.

Keywords: EJC

87. FMRP inhibits translation elongation independent of G-quadruplexes

MaKenzie R. Scarpitti (The Biomedical Sciences Graduate Program, Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University), Michael G. Kearse (The Biomedical Sciences Graduate Program, Department of Biological Chemistry and Pharmacology, Center for RNA Biology, The Ohio State University)

Abstract:
Loss of expression of fragile X mental retardation protein (FMRP) causes fragile X syndrome, the leading form of inherited intellectual disability and the most common monogenic cause of autism spectrum disorders. FMRP is an RNA-binding protein that controls neuronal mRNA localization and translation. How FMRP controls translation remains largely unclear but it may use multiple mechanisms to target different steps of translation. Most notably, FMRP is thought to inhibit translation elongation on target transcripts that contain RNA G-quadruplex (G4) structures within the coding sequence. Here we directly test this model and report that FMRP inhibits translation elongation independent of G4s in the mRNA coding sequence. Furthermore, we define that the RGG box motif together with its natural C-terminal domain forms a non-canonical RNA-binding domain (ncRBD) that enables FMRP to bind mRNA independently of sequence or structure. Only when FMRP is bound to mRNA through this ncRBD does FMRP inhibit translation. Furthermore, the ncRBD of FMRP stalls elongating ribosomes on transcripts and thereby promotes accumulation of heavy polysomes. Together, this work shifts our understanding of how FMRP represses translation elongation and supports a model where repression is driven by local FMRP concentration rather than mRNA sequence.

Keywords: RNA binding protein, mRNA, ribosomes

89. Chemical tools to incorporate a trimethyl guanosine cap onto model RNAs and to study oxidative damage to RNA

Deepak Shrestha (Chemistry, Wayne State University ), Evan Jones (Chemistry, Wayne State University ), Dr. Mel Bedi (Pharmacy, Wayne State University), Dr. Christine Chow (Chemistry, Wayne State University), Dr. Amanda Bryant-Friedrich (Pharmacy, Wayne State University )

Abstract not available online - please check the booklet.

90. Title not available online - please see the booklet.

Jalal Siddiqui (Comprehensive Cancer Center, The Ohio State University), Wayne Miles (Cancer Biology and Genetics, The Ohio State University)

Abstract not available online - please check the booklet.

91. The role of a nuclear-enriched long non-coding RNA in hypoxia response and cancer progression

You Jin Song (Department of Cell and Developmental Biology, UIUC), Prasanth Kannanganattu (Department of Cell and Developmental Biology, UIUC)

Abstract not available online - please check the booklet.

92. RESC8 and RESC14 cooperate to mediate RESC function and dynamics during trypanosome RNA editing

Katherine Sortino (Department of Microbiology & Immunology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203.), Brianna Tylec (Department of Microbiology & Immunology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203.), Natalie M. McAdams (Department of Microbiology & Immunology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203.), Runpu Chen (Department of Microbiology & Immunology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203.), Yijun Sun (Department of Microbiology & Immunology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203.), Laurie K. Read (Department of Microbiology & Immunology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203.)

Abstract:
Uridine insertion/deletion RNA editing is unique to kinetoplastid organisms and essential for their survival and virulence. This intricate process requires the action of several multiprotein complexes and small trans-acting RNAs called guide RNAs. The RNA editing holoenzyme comprises three dynamically interacting complexes: RECC (RNA Editing Catalytic Complex), RESC (RNA Editing Substrate Binding Complex), and REH2C (RNA Editing Helicase 2 Complex). Previous studies in Trypanosoma brucei showed that RESC components, RESC14 and RESC8, are critical in maintaining normal protein-protein and protein-RNA interactions within the editing holoenzyme. To further clarify the role of these proteins and the RESC complex in RNA editing, we performed high-throughput sequencing of ATP synthase subunit 6 (A6) mRNA in RESC14, RESC8, and RESC13 RNAi cells, and compared the phenotypes of these knockdown cell lines. We measured a highly significant overlap between exacerbated pause sites arising on A6 mRNA in cells depleted of RESC14 and RESC8, suggesting an overlapping function between these proteins during editing progression. In contrast, A6 mRNA from RESC13 depleted cells exhibited different pause sites, indicative of a function distinct from that of RESC14/8. Additional features of editing progression, such as the lengths of mis-edited junctions, also differed between RESC14/8 and RESC13, supporting their specific functions and suggesting that both RESC14 and RESC8 are needed for junction formation on A6 mRNA. To probe the biochemical basis of the RESC14/8 interdependence, we performed Native PAGE analysis and showed that RESC8 is not incorporated into large editing complexes in the absence of RESC14. Co-immunoprecipitation assays also identified an RNA inhibited interaction between the RESC14 and RESC8. Together, our findings suggest that RESC14 and RESC8 rely on each other for necessary protein and RNA rearrangements during editing progression. Our working model is presented.

Keywords: Trypanosome, RNA editing

93. Screening of Group IIC Intron IEP Tags for RNP Kinetic Studies

Sarah A. Starcovic (Department of Biochemistry, West Virginia University), Olivia P. Carpenter (Department of Biochemistry, West Virginia University), Aaron R. Robart (Department of Biochemistry, West Virginia University)

Abstract:
Group II introns are self-splicing ribozymes that catalyze intron self-splicing reactions. Splicing reactions occur through two competing reactions, a non-physiological hydrolytic reaction, and a branched splicing reaction that results in the formation of intron lariats. Group II introns often encode an intron-encoded protein (IEP) which assists splicing. A primitive sub-type of these genetic elements, known as IIC introns, require the IEP to efficiently form intron lariat products. Interestingly, IIC lack many RNA secondary structural features associated with ribozyme catalyzed lariat formation, suggesting that the IEP compensates for the “missing” RNA structure. Due to this ribonucleoprotein (RNP) dependence, IIC introns offer a unique opportunity to understand how self-splicing ribozymes transitioned into the RNP dependent splicing machines found in eukaryotic cells. Here, we report the expression and purification of several different affinity tags attached to the IEP. Our immediate goals are to use these affinity tags to tether the IEP to surface plasmon resonance (SPR) chips to study binding and dissociation kinetics. Each tagged IEP was purified, checked for splicing activity, and if applicable, an RNP pulldown was performed. Use of multiple purification tags will also aid our ongoing RNP purification efforts for cryo-EM structural studies.

Keywords: ribonucleoprotein, SPR, IEP

94. A Transcriptome Wide Study to Elucidate co-Translational and Translation-independent mRNA Decay Kinetics

Brittany Stawicki (Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, OH), Eckhard Jankowsky (Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, OH)

Abstract:
Regulation of eukaryotic gene expression is controlled by changes in mRNA levels. It is known that mRNAs can be degraded co-translationally as well as independent of translation, but it is unclear whether and how these decay pathways are balanced. Current approaches to measure mRNA decay are unable to distinguish between translation-dependent and independent degradation. Therefore, we set out to directly measure co-translational and translation-independent mRNA decay kinetics on a transcriptome-wide scale. We perform SLAMseq 4-thiouridine (4sU) labeling of human HCT116 cells under pulse chase conditions, followed by Quantseq analysis of mRNA 3’ends to track metabolically labeled RNA populations over time in the overall and translating mRNA pool. This approach overcomes limitations from current metabolic labeling procedures; including high label concentrations, long labeling times, and bias introduced by affinity pulldown. While optimizing 4sU labeling and pulse-chase conditions, we noted that commonly used labeling conditions are associated with adverse effects on translation, likely distorting present decay kinetics. Our optimized conditions minimize the effects of metabolic labeling and pulse-chase on translation. We are currently sequencing and analyzing obtained NGS libraries. We anticipate our approach measuring co-translational and translation-independent mRNA decay rates to provide kinetic parameters for key stages in mRNA metabolism, building on existing data. Our approach will supply quantitative insights into the regulation of RNA metabolism and aid in establishing a foundation for systematic analyses of diseases associated with changes in RNA metabolism.

Keywords: RNA Metabolism, Decay

95. Experimental approaches to identify novel RNAs with tertiary structure

Nishanti Sudhakar (BMMB, Penn State University), Dr. Philip C. Bevilacqua (Chemistry, Penn State University)

Abstract not available online - please check the booklet.

96. Structural basis for the context-specific action of classic peptidyl transferase inhibitors

Egor A. Syroegin (Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA), Laurin Flemmich (Institute of Organic Chemistry, University of Innsbruck, Center of Molecular Biosciences Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria), Dorota Klepacki (Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA), Nora Vazquez-Laslop (Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA), Ronald Micura (Institute of Organic Chemistry, University of Innsbruck, Center of Molecular Biosciences Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria), Yury S. Polikanov (Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA)

Abstract:
Ribosome-targeting antibiotics serve both as powerful antimicrobials and as tools for studying the ribosome. The ribosomal catalytic site, the peptidyl transferase center (PTC), is targeted by a large number of various drugs. The classical and best-studied PTC-acting antibiotic chloramphenicol, as well as the newest clinically significant linezolid, were considered indiscriminate inhibitors of every round of peptide bond formation, presumably inhibiting protein synthesis by stalling ribosomes at every codon of every gene being translated. However, it was recently discovered that chloramphenicol or linezolid, and many other PTC-targeting drugs, preferentially arrest translation when the ribosome needs to polymerize particular amino acid sequences. The molecular mechanisms and structural bases that underlie this phenomenon of context-specific action of even the most basic ribosomal antibiotics, such as chloramphenicol, are unknown. Here we present high-resolution structures of ribosomal complexes, with or without chloramphenicol, carrying specific nascent peptides that support or negate the drug action. Our data suggest that specific amino acids in the nascent chains directly modulate the antibiotic affinity to the ribosome by either establishing specific interactions with the drug molecule or obstructing its placement in the binding site. The model that emerged from our studies rationalizes the critical importance of the penultimate residue of a growing peptide for the ability of the drug to stall translation and provides the first atomic-level understanding of context specificity of antibiotics that inhibit protein synthesis by acting upon the PTC

Keywords: antibiotics, ribosome, chloramphenicol

97. Analysis of HTLV-1 Gag interactome and characterization of its chaperone activity

Yu-Ci Syu (Molecular, Cellular, and Developmental Biology Graduate Program, Center for RNA Biology, Center for Retrovirus Research, The Ohio State University), Joshua Hatterschide (Department of Chemistry and Biochemistry, Center for RNA Biology, Center for Retrovirus Research, The Ohio State University), Yingke Tang (Department of Chemistry and Biochemistry, Center for RNA Biology, Center for Retrovirus Research, The Ohio State University), Amanda R. Panfil (Molecular, Cellular, and Developmental Biology Graduate Program, Department of Veterinary Biosciences, Center for Retrovirus Research, The Ohio State University), Patrick L. Green (Molecular, Cellular, and Developmental Biology Graduate Program, Department of Veterinary Biosciences, Center for Retrovirus Research, The Ohio State University), Karin Musier-Forsyth (Molecular, Cellular, and Developmental Biology Graduate Program, Department of Chemistry and Biochemistry, Center for RNA Biology, Center for Retrovirus Research, The Ohio State University)

Abstract:
Human T-cell leukemia virus type 1 (HTLV-1) is the only oncogenic human retrovirus discovered to date. All retroviruses use a host cell tRNA to prime RT. The 3' end of the tRNA anneals to an 18-nucleotide region of the genome known as the primer binding site (PBS). In HTLV-1, the PBS is complementary to human tRNAPro. Structure-probing of the genomic RNA revealed the PBS is embedded in a highly-structured hairpin.1 HTLV-1 nucleocapsid (NC), matrix (MA), and even the HIV-1 NC chaperone protein are incapable of annealing tRNAPro to the stable PBS domain in vitro. We hypothesize that HTLV-1 Gag may have more robust chaperone activity and that a cellular co-factor may be required to facilitate primer tRNA annealing. We successfully purified recombinant HTLV-1 Gag for the first time and performed fluorescence anisotropy RNA-binding assays, as well as primer-annealing assays. In direct-binding assays to HTLV-1 5' UTR-derived RNAs, we found that HTLV-1 Gag displayed stronger RNA-binding affinity (Kd ~0.2-6 uM) than HTLV-1 NC (~3 uM) or MA (~2-8 uM). As expected, mutant HTLV-1 Gag with deletion of the highly acidic 29 residues at the C-terminus displayed significantly better RNA-binding activity (Kd ~0.2-1.5 uM) than WT HTLV-1 Gag (~0.5-6 uM). In annealing assays, mutant HTLV-1 Gag chaperoned the annealing of tRNAPro to the PBS more efficiently than WT HTLV-1 Gag. To identify potential interacting partners of HTLV-1 Gag in cells, we performed affinity tagging/purification mass spectrometry (AP-MS). Two significant AP-MS hits, RPL7 and YBX1, were further validated by reciprocal co-IP studies in both HEK293T cells and chronically HTLV-1 infected MT-2 cells. RPL7 is a ribosomal protein that has been reported to be packaged into HIV-1 virions and interact with the NC domain of HIV-1 Gag to increase chaperone activity.2 YBX1 is a transcription factor and an RNA-binding protein shown to interact with tRNA-derived fragments to mediate the degradation of oncogene transcripts.3 Current studies are focused on preparing recombinant RPL7 and investigating whether it enhances HTLV-1 Gag’s chaperone activity. Taken together, these studies will lead to mechanistic insights that could be exploited for new therapeutic strategies.

References:
1. Wu, W. et al. Human T-cell leukemia virus type 1 Gag domains have distinct RNA-binding specificities with implications for RNA packaging and dimerization. J Biol Chem 293, 16261-16276, (2018).
2. Mekdad, H. E. et al. Characterization of the interaction between the HIV-1 Gag structural polyprotein and the cellular ribosomal protein L7 and its implication in viral nucleic acid remodeling. Retrovirology 13, 54, (2016).
3.Goodarzi, H. et al. Endogenous tRNA-Derived Fragments Suppress Breast Cancer Progression via YBX1 Displacement. Cell 161, 790-802, (2015).

Keywords: HTLV-1, tRNA, RPL7

99. ELeFHAnt: An automated tool for harmonization and annotation of single cell RNA-seq data

Konrad Thorner (Cincinnati Childrens Hospital Medical Center, Developmental Biology), Praneet Chaturvedi (Cincinnati Childrens Hospital Medical Center, Developmental Biology)

Abstract:
Single cell sequencing has become an important method for understanding biological systems at an increasingly granular level. For single cell RNA-seq data specifically, one of the primary questions is using the transcriptome to determine cell identity. It is common to visualize such data and find clusters of cells with similarity in gene expression, but assigning each cluster a cell type is a much more open-ended task. Taking advantage of publicly available, annotated datasets in combination with supervised learning is a powerful approach for addressing this question. Ensemble Learning for Harmonization and Annotation of Single Cells (ELeFHAnt) provides an easy-to-use R package for users to annotate clusters of single cells, harmonize labels across single cell datasets to generate a unified atlas, and infer relationships among cell types between two datasets. It provides users with the flexibility of choosing between random forest and SVM (Support Vector Machine) based classifiers or letting ELeFHAnt apply both in combination to make predictions. ELeFHAnt is available on GitHub: https://github.com/praneet1988/ELeFHAnt

Keywords: machine learning, annotation, single cell RNA-seq

100. Investigation of RNA 3D structure & small molecule interaction by a multidisciplinary approach

Elizabeth D. Tidwell (Biophysics University of Michigan), Anna Anders (Chemistry University of Michigan), Varun Gadkari (Chemistry University of Michigan), Aaron T Frank (Biophysics University of Michigan), Brandon T Ruotolo (Chemistry University of Michigan), Markos Koutmos (Biophysics and Chemistry University of Michigan)

Abstract not available online - please check the booklet.

101. Alternative transcription start sites (aTSS): a mechanism for post-transcriptional regulation of C. albicans filamentation

Melissa A. Tosiano (Biological Sciences, Carnegie Mellon University ), Gemma E. May (Biological Sciences, Carnegie Mellon University ), Derek Wang (Biological Sciences, Carnegie Mellon University ), C. Joel McManus (Biological Sciences, Carnegie Mellon University )

Abstract:
While Candida albicans lives as a commensal organism in healthy humans, it can cause candidiasis in immunocompromised individuals. Its ability to detect and respond to stressors in diverse physiological microenvironments is required for pathogenicity1. In response to stress, C. albicans undergoes filamentation - shifting from yeast to virulent hyphae2. Post-transcriptional regulation contributes to eukaryotic stress responses and may play a role in C. albicans filamentation. Indeed, the TLs of UME6 and EFG1, two C. albicans filamentation inducing transcription factors, possess long complex 5’ TLs hypothesized to modulate translation3,4. Alternative transcription start sites (aTSSs) create transcript leaders with substantially different translation efficiencies in other fungi5 and can lead to N-terminally truncated proteins and / or decreased expression through nonsense-mediated mRNA decay (NMD). This suggests a role for aTSSs during C. albicans filamentation. Using public C. albicans TL sequencing data, we evaluated the locations of m7G capped transcription start site (TSS) peaks. We found many ORFs possess at least one alternative TSS, a fraction of which had an aTSS within the CDS. Additionally, we identified 575 ORFs with long TLs likely to contain cis-regulatory elements. Using 5’ end sequencing we generated TL maps from C. albicans before and 90 minutes after exposure to filament inducing stress. In our future work, we will quantify TE of differential TLs transcribed in the yeast and hyphal phenotypes.

References:
1. Brown, A. J. P. et al. Stress adaptation in a pathogenic fungus. J. Exp. Biol. 217, 144–155 2014.
2. Thompson, D. S., et al. Coevolution of morphology and virulence in Candida species. Eukaryot. Cell 10, 1173–1182 2011.
3. Desai, P. R. et al. The 5’ Untranslated Region of EFG1 Transcript Promotes Its Translation To Regulate Hyphal Morphogenesis in C. albicans. mSphere. 3, 1–15 2018.
4. Childers, D. S., et al. A 5’ UTR-mediated translational efficiency mechanism inhibits the C. albicans morphological transition. Mol. Microbiol. 92, 570–585 2014.
5. Arribere, J. A. & Gilbert, W. V. Roles for TLs in translation and mRNA decay revealed by transcript leader sequencing. Gen. Res. 23, 977–987 2013.

Keywords: C albicans, aTSS, post-transcriptional regulation

102. The role of circular RNA in Parkinson’s disease etiology

Megan L. Van Horn (Carnegie Mellon University), Erika Kim (Carnegie Mellon University), Anna M. Kietrys (Carnegie Mellon University)

Abstract:
The world’s population is ageing, and this phenomenon correlates with a growing number of people diagnosed with neuronal disorders1. One of these late-onset diseases is Parkinson’s disease (PD), the second most common neurodegenerative disorder worldwide. Parkinsonism affects more than 1 million people in North America and over 60,000 new cases are identified every year2. Despite the effort of many research groups, very little is known about where and why neuronal degradation starts. To propose and develop an effective therapy for PD, we first need to understand the etiology of neurodegeneration.

Our group targets the problem of RNA-driven neurodegeneration and proposes new therapeutic approaches. Utilizing a high-throughput differential analysis of PD transcriptomes, we have identified circular RNA (circRNA) molecular markers that correlate with ageing and parkinsonism. Circular RNAs, because of their extended life span3 and a versatile interaction with small RNAs4,5 and proteins, are a strong candidate as cellular regulators of neurodegeneration3,5.

To identify potential novel circRNAs correlated with Parkinson’s disease stages, we analyzed multiple RNA-seq data sets collected from human specimens. The first set of data (control = 44, PD = 29) has shown a circRNA originating from chromosome 1 in the NBPF1 coding region. This underexpressed chr1 circRNA was found to have 97 potential miRNA binding sites and multiple associated RNA-binding proteins. These include EIF4A3, FMRP, and SFRS1, which are all linked to translation and splicing regulation, indicating this circRNA as a potential translation or alternative splicing influencer. In the same data set, PD samples showed significant overexpression of a mitochondrial circRNA originating from the COX3 coding region. This previously unknown circRNA possesses 21 potential miRNA binding sites. Both of these circRNAs have been experimentally found to be expressed in HEK293T and U87MG cells. We believe these identified circRNAs may trigger the neuron degeneration process and in the future be utilized as therapeutic targets.

References:
1. Tran J, Anastacio H, Bardy C. npj Park Dis. 2020. doi:10.1038/s41531-020-0110-8
2. Marras C, Beck JC, Bower JH, et al. npj Park Dis 2018. doi:10.1038/s41531-018-0058-0
3. Piwecka M, Glažar P, Hernandez-Miranda LR, et al. Science 2017. doi:10.1126/science.aam8526
4. Hansen TB, Jensen TI, Clausen BH, et al. Nature 2013. doi:10.1038/nature11993
5. Memczak S, Jens M, Elefsinioti A, et al. Nature 2013. doi:10.1038/nature11928

Keywords: circular RNA, Parkinsons disease, neurodegeneration

103. Heat restores full-length SMN transcripts: promise for splice-augmenting therapies in SMA

Akila S. Venkataramany (Biomedical Sciences Graduate Program and Medical Scientist Training Program, The Ohio State University), Catherine E. Dominguez (Molecular, Cellular, & Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University), David Cunningham (Center for Childhood Cancer and Blood Diseases, Research Institute at Nationwide Childrens Hospital), Dawn S. Chandler (Molecular, Cellular & Developmental Biology Graduate Program and The Center for RNA Biology, OSU ; Center for Childhood Cancer and Blood Diseases, Research Institute at Nationwide Childrens Hospital)

Abstract not available online - please check the booklet.

104. A computational pipeline for RNA 8-oxoguanine accumulation caused by oxidative stress

Derek Wang (Carnegie Mellon University, Department of Biological Sciences), C. Joel McManus (Carnegie Mellon University, Department of Biological Sciences)

Abstract not available online - please check the booklet.

105. Effects of an alternative rRNA helix on 30S subunit biogenesis.

Benjamin R. Warner (Department of Microbiology, The Ohio State University), Kurt L. Fredrick (Department of Microbiology, The Ohio State University)

Abstract not available online - please check the booklet.

107. Structural and kinetic studies of a minimal protein-only RNase P

Catherine A. Wilhelm (Department of Chemistry at the University of Michigan), Kipchumba J. Kaitany (Department of Chemistry at the University of Michigan), Leena Mallik (Center for Structural Biology at the University of Michigan), Markos Koutmos (Department of Chemistry at the University of Michigan)

Abstract:
RNase P enzymes are responsible for pre-tRNA processing and maturation [1] across all domains of life. While the vast majority of RNase P enzymes require an RNA co-factor, Protein-Only RNase P enzymes (PRORPs) are found in human mitochondria and all tRNA-encoding compartments of plants[2]. PRORPs canonically consist of a pentatricopeptide repeat (PPR) RNA recognition domain, as well as a catalytic metallonuclease domain[3]. PRORPs were long thought to be the minimal RNase P construct, until the discovery of a further truncation in Aquifex aeolicus consisting of only the metallonuclease domain[4]. As these Homologs of Aquifex aeolicus RNase P (HARPs) lack the canonical PPR substrate recognition domain, we aim to characterize the substrate recognition and specificity of HARP enzymes utilizing a combination of electrophoretic mobility shift binding assays (EMSAs), single turnover assays, x-ray crystallography, and cryo-EM. Interestingly, our current structural data suggests that HARPs form a hexamer ring of six homodimers with twelve hypothetical active sites. Thus, we seek to determine the binding stoichiometry of HARPs, with preliminary data indicating that HARPs undergo multiple binding events and can specifically cleave multiple species of pre-tRNA without off-target activity. Furthermore, we seek to determine the observed rate constant of pre-tRNA cleavage and observe how active site mutants contribute to catalysis and substrate binding.

References:
[1] Altman and Smith. Nature 1971; 233:35-39.
[2] Daniels et al. RNA 2019; 25:286-291.
[3] Gobert et al. Nat Comm 2013; 4:1353.
[4] Nickel et al. PNAS 2017; 114(42):11121-11126.

Keywords: tRNA processing, RNA-binding proteins, protein structure

108. Title not available online - please see the booklet.

Sydney Willey (Department of Neuroscience Student, The Ohio State University), Yufeng Liang (Department of Chemistry and Biochemistry, The Ohio State University), Yu-Chieh Chung (Department of Biological Chemistry and Pharmacology, The Ohio State University), Dennis Bong (Department of Chemistry and Biochemistry, The Ohio State University), Li-Chun Tu (Department of Biological Chemistry and Pharmacology, The Ohio State University)

Abstract not available online - please check the booklet.

109. The NiCo riboswitch as an iron-responsive riboswitch: a new mechanism of bacterial iron regulation

Jiansong Xu (Department of Chemistry, Penn State University), Joseph Cotruvo (Department of Chemistry, Penn State University)

Abstract not available online - please check the booklet.

110. Development of tools for TERRA imaging

MengXu (Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University), Tafadzwa Chigumira (Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University), Ziheng Chen (Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University), Jason Tone (Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University), Huaiying Zhang (Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University)

Abstract not available online - please check the booklet.

111. Molecular mechanisms that mediate A-to-I RNA editing in vivo

Boyoon Yang (Molecular and Cellular Biochemistry, Indiana University Bloomington), Heather Hundley (Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN)

Abstract not available online - please check the booklet.

112. Dynamics of RNA Conformational Sampling by the SMK box riboswitch

Haoyun Yang (Department of chemistry and biochemistry, OSU), Mark Foster (Department of Chemistry and Biochemistry, OSU), Tina Henkin (Department of Microbiology, OSU)

Abstract:
The conformational dynamics of non-coding RNAs are coupled to vital cellular processes such as metabolite sensing, site specific RNA catalysis and the hierarchy of ordered ribonucleoprotein assemblies. However, the free energy landscapes that enable RNA functional conformational dynamics remain poorly understood. To fill this knowledge gap, we study a model system, the SMK box, S-adenosylmethionine (SAM) metabolite-binding/SAM-III riboswitch RNA, a compact RNA that undergoes ligand-mediated conformational changes to alter ribosome binding and thereby regulate translation1,2. Previously, X-ray crystallography analysis of the SMK aptamer bound to SAM established the structure of the ligand-bound, repressed state3. NMR, mutagenesis and chemical probing data suggest that in the absence of SAM, SMK is in equilibrium between an alternative fold (ISO) that has no obvious ligand binding site and a ligand binding-competent state (PRIMED)4,5. To understand how the SMK box riboswitch is able to undergo conformational fluctuations between alternative structures that enable its function we use stopped-flow measurements of SAM binding to 2-aminopurine labeled RNA to discriminate between conformational selection and induced-fit mechanisms of ligand binding. These experiments also quantify the thermodynamic barriers between conformations and provide structural insights into those rate-liming steps. Heteronuclear NMR spectroscopy is applied to determine the structures of the low-energy states, and the pathways connecting them.

References:
1. Fuchs RT, Grundy FJ, Henkin TM. S-adenosylmethionine directly inhibits binding of 30S ribosomal subunits to the SMK box translational riboswitch RNA. Proc Natl Acad Sci U S A. 2007;104(12)
2. Fuchs RT, Grundy FJ, Henkin TM. The S(MK) box is a new SAM-binding RNA for translational regulation of SAM synthetase. Nat Struct Mol Biol. 2006;13(3)
3. Lu C, Smith AM, Fuchs RT, et al. Crystal structures of the SAM-III/S(MK) riboswitch reveal the SAM-dependent translation inhibition mechanism. Nat Struct Mol Biol. 2008;15(10)
4. Lu C, Smith AM, Ding F, Chowdhury A, Henkin TM, Ke A. Variable sequences outside the SAM-binding core critically influence the conformational dynamics of the SAM-III/SMK box riboswitch. J Mol Biol. 2011;409(5)
5. Wilson RC, Smith AM, Fuchs RT, Kleckner IR, Henkin TM, Foster MP. Tuning riboswitch regulation through conformational selection. J Mol Biol. 2011;405(4)

Keywords: Riboswitch , Dynamics , NMR

113. Towards quantitative measurements of global RNA-protein binding patterns in cells.

Soon Yi (Department of Biochemistry and Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH), Eckhard Jankowsky (Department of Biochemistry and Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH)

Abstract not available online - please check the booklet.

114. Mammalian UPF3A and UPF3B activate NMD independently of their EJC binding

Zhongxia Yi (Center for RNA Biology, Department of Molecular Genetics, the Ohio State University), Rene M Arvola, Sean Myers, Corinne N Dilsavor, Rabab Abu Alhasan, Bayley N Carter, Robert D Patton, Ralf Bundschuh, Guramrit Singh

Abstract:
Nonsense-mediated mRNA decay (NMD) is governed by the three conserved factors - UPF1, UPF2 and UPF3. While all three are required for NMD in yeast, UPF3B is dispensable for NMD in mammals, with its paralog UPF3A suggested to only weakly activate or even repress NMD due to its weaker binding to the exon junction complex (EJC). Here we characterize the UPF3B-dependent and -independent NMD in human cell lines knocked-out of one or both UPF3 paralogs. We show that in human colorectal cancer HCT116 cells, EJC-mediated NMD can operate in UPF3B-dependent and -independent manner. While UPF3A is almost completely dispensable for NMD in wild-type cells, it strongly activates EJC-mediated NMD in cells lacking UPF3B. Surprisingly, this major NMD branch can operate in UPF3-independent manner questioning the idea that UPF3 is needed to bridge UPF proteins to the EJC during NMD. Complementation studies in UPF3 knockout cells further show that EJC-binding domain of UPF3 paralogs is not essential for NMD. Instead, the conserved mid domain of UPF3B, previously shown to engage with ribosome release factors, is required for its full NMD activity. Altogether, UPF3 plays a more active role in NMD than simply being a bridge between the EJC and the UPF complex.

Keywords: Nonsense-mediated mRNA decay, UPF3, Exon Junction Complex

115. Natural variation in the plant polyadenylation complex

Lichun Zhou (Department of Plant and Soil Sciences, University of Kentucky), Kai Li (Department of Plant and Soil Sciences, University of Kentucky), Arthur G. Hunt (Department of Plant and Soil Sciences, University of Kentucky)

Abstract:
Messenger mRNA polyadenylation, the process wherein the primary RNA polymerase II transcript is cleaved and a poly(A) tract added, is mediated by a complex (the polyadenylation complex, or PAC) that consists of between 15 and 20 subunits. In plants, as many as fifteen subunits have been identified by sequence comparisons with the mammalian and yeast polyadenylation complexes, and some additional plant-specific subunits identified using genetic approaches. While analogies with the mammalian and yeast complexes have provided a good working understanding of the plant PAC, several aspects of the plant PAC raise interesting questions. For example, one subunit of the PAC, CPSF30 (or CPSF4), is an essential subunit in mammals and yeast, but entirely dispensable for plant growth and development [1]. To gain further insight into novelty in the plant PAC, we have begun a study of variation in subunits of the complex. For this, we drew upon a database of naturally-occurring variation in numerous geographic isolates of Arabidopsis thaliana [2]. Most of the subunits of the Arabidopsis CPSF and CstF complexes show patterns of variability that are consistent with their roles as essential proteins. However, for some subunits, the distributions of non-synonymous mutations suggest possible ancillary or regulatory functions apart from those needed for cleavage and polyadenylation. For a small subset of PAC subunits, the patterns of variability are characterized by relatively high ratios of non-synonymous/synonymous substitutions and the occurrence of premature stop codons in some Arabidopsis accessions. These features are most prominent in subunits encoded by gene families, and strongly suggest that some members of these families have lost function and are instead pseudogenes. In addition, some PAC subunits – isoforms of Pcf11, CFIm68, PAP, and FIP1 – have patterns of variation consistent with selection for new or altered function. We propose that the patterns of natural variation seen in Arabidopsis PAC subunits reflect both core functionality and regulatory interactions important for differential usage of poly(A) sites during growth and development.

References:
1. Hunt AG. mRNA 3' end formation in plants: Novel connections to growth, development and environmental responses. Wiley Interdiscip Rev RNA. 2020;11(3):e1575.
2. Genomes Consortium. 1,135 Genomes Reveal the Global Pattern of Polymorphism in Arabidopsis thaliana. Cell. 2016;166(2):481-91.

Keywords: polyadenylation, Arabidopsis, evolution

116. Antimicrobial Peptides that Target RNA: Applied in a Flexible Laboratory Curriculum for Educating Interdisciplinary Scientists

Teresa A. Zimmerman (Department of Chemistry, Wayne State University, Detroit, MI), Rabiul Islam (Department of Chemistry, Wayne State University, Detroit, MI), Christine S. Chow (Department of Chemistry, Wayne State University, Detroit, MI)

Abstract:
Highly skilled and interdisciplinary scientists with a strong background in wet and dry laboratory techniques are increasingly in demand. The techniques are often taught separately with the class curriculum focusing on behind the bench, wet techniques, or computational, dry techniques. This approach creates a gap in application from one technique to another. We have developed a combinational laboratory curriculum that applies bioinformatic analysis to molecular biology techniques while allowing students to address issues such as antibiotic resistance and RNA targeting. This curriculum focuses on the proline-rich antimicrobial peptide oncocin but can be applied to other antimicrobial peptides. Students are introduced to software and online resources that aid in primer design for cloning experiments. Using non-template PCR provides a cost-effective protocol that allows students to design their own derivative of a naturally occurring antimicrobial peptide, which will be coded for in their primers. The antimicrobial activities of the designed peptides are then tested in vivo to determine bacterial killing efficiency. Using derivatives of natural products that have a known target such as rRNA opens the door to other bioinformatic analyses such as multiple sequence alignments (MSA) and computational methods such as docking. As an example, oncocin binds to the bacterial ribosome to prevent translation; an MSA of different bacterial species was performed to determine the percent conservation of the RNA binding domain. Simulated docking studies can aid in improving oncocin derivatives for increased antimicrobial activity. Our curriculum design allows educators to offer some aspects of the laboratory class remotely and allows flexibility in the schedule if unforeseen circumstances arise.

References:
1. Muthunayake, N. S.; Islam, R.; Inutan, E. D.; Colangelo, W.; Trimpin, S.; Cunningham, P. R.; Chow, C. S., Expression and In Vivo Characterization of the Antimicrobial Peptide Oncocin and Variants Binding to Ribosomes. Biochemistry 2020, 59 (36), 3380-3391.

Keywords: antibiotics, bioinformatics, interdisciplinary