Author Archives: Joanne Lim

Links to entries

DocuLearn 1:https://blogs.ubc.ca/joannebiol463/2018/09/11/doculearn1-my-goals-and-self-regulated-learning/

DocuLearn 2:https://blogs.ubc.ca/joannebiol463/2018/09/20/doculearn2/

DocuLearn 3: https://blogs.ubc.ca/joannebiol463/2018/12/03/doculearn3/

DocuLearn 4: https://blogs.ubc.ca/joannebiol463/2018/12/03/doculearn4/

DocuLearn 5: https://blogs.ubc.ca/joannebiol463/2018/12/03/doculearn-5/

Top 4 Assignments

caste study stage 1: https://blogs.ubc.ca/joannebiol463/2018/12/03/case-study-stage-1/

Lonfat paper: https://blogs.ubc.ca/joannebiol463/2018/12/03/cooperative-assignment-lonfat-et-al/

Wang paper: https://blogs.ubc.ca/joannebiol463/2018/12/04/top4-wang-et-al/

Quiz 4: https://blogs.ubc.ca/joannebiol463/2018/12/04/top4-quiz-4/

Links to entries

If I were a developmental biologist: https://blogs.ubc.ca/joannebiol463/2018/09/11/if-i-were-a-developmental-biologist/

Project question: https://blogs.ubc.ca/joannebiol463/2018/09/29/final-project-question/

project outline: https://blogs.ubc.ca/joannebiol463/2018/12/03/final-project-outline/

project draft: https://blogs.ubc.ca/joannebiol463/2018/12/03/project-draft/

final final project: https://blogs.ubc.ca/joannebiol463/2018/12/04/final-final-project/

annotated bibliography: https://blogs.ubc.ca/joannebiol463/2018/12/03/annotated-bibliography/

how you went about developing your project: https://blogs.ubc.ca/joannebiol463/2018/12/03/pathway-to-our-final-final-project/

Final Final Project

Long noncoding RNA UCA1: Function in Cisplatin Resistance in Neuroblastoma Cell Lines

Shannah Fisher and Joanne Lim

Background

Neuroblastoma is a childhood malignancy in the sympathetic nervous system and accounts for 15% of all deaths in pediatric cancer patients (Piskareva et al, 2015). Cisplatin, a platinum based cytotoxic drug, is one of the most common chemotherapy agents used to treat neuroblastoma (Dasari and Tchounwou, 2014). However, a large proportion of patients are resistant to cisplatin-based therapies either at the beginning of treatment or after ongoing exposure to treatment (Galluzzi, 2012). In order to effectively treat cancer, it is important to understand the mechanism of cancer resistance and find ways to prevent their effect (Ayers and Vandesompele, 2017).

 

Cisplatin resistance is not only found in neuroblastoma but also in other cancers, including breast, cervical, lung and bladder cancer (Dasari and Tchounwou, 2014). Mechanisms of cisplatin resistance include change in the signaling pathways, silencing of certain genes by miRNA, changes to the cell cycle, development of an efflux system, and DNA repair (Chen, 2017). Several studies have also revealed that long non-coding RNAs (lncRNAs) are involved in chemoresistance to cisplatin by interacting with histone modification tools and with other chromatin regulatory factors (Guttman et al., 2011).

 

Long non-coding RNAs (lncRNAs) are evolutionary conserved gene transcripts of over 200 nucleotides that do not encode proteins (Ponting et al., 2009). They are involved in regulation of gene expression at the transcriptional, post-transcriptional, and translational levels (Moran et al., 2012) either by directly interacting with a target gene (cis-acting) or by interacting with transcription factors (trans-acting) (Ponting et al., 2009). A variety of lncRNAs such as HOTAIR, MALAT1, and UCA1 have been found to play an important role in carcinogenesis, metastasis, prognosis and treatment. In particular, Urothelial cancer-associated 1 (UCA1), a lncRNA that has a regulatory function in proliferation of cells, has been found to play a major role in development of cisplatin resistance in bladder cancer, breast cancer, hepatocellular carcinoma, ovarian cancer, and tongue squamous cell carcinoma (Wang et al., 2017).

 

Studies looking at cisplatin resistance in numerous cancers have found a relationship between lncRNA UCA1 and changes in expression levels of genes that are involved in the cells susceptibility to treatment (Wang et al, 2017). This includes a study performed by Wang et al, where expression levels of 42 genes were found to change by at least two-fold in the presence of UCA1 in bladder cancer, including an upregulation of Wnt signaling pathway member 6 (Wnt6), CYP1A1 (a cytochrome) and AURKC (kinase) and a downregulation of methyl‐CpG binding domain protein 3 (MBD3) and SR (serine/arginine‐rich) protein‐specific kinase 1 (SRPK1). These results are verified by several other studies looking further into the effects of UCA1 on gene expression in bladder cancer and ovarian cancer (Fan et al, 2014; Wang et al, 2015).

Relevance

The function of lncRNA UCA1 in cisplatin resistance has never been studied  neuroblastoma, posing a gap in knowledge that could be vital to finding effective treatment methods for patients who are resisting this drug. Although UCA1 has been studied in other cancers, we cannot assume that the effect of UCA1 will be conserved across all cancer types, including neuroblastoma. For example, lncRNA MALAT1 functions as a tumor suppressor gene in breast cancer (Eastlack, 2018), but as a promoter of tumor growth and metastasis in oral squamous cell carcinoma (Zhou, 2015). Therefore, it is possible that lncRNA UCA1 may take a unique function depending on the cancer cell type.

 

Therefore, we propose a study that will determine the function of lncRNA UCA1 in cisplatin resistance specific to neuroblastoma cell lines and identify the changes in expression levels of various genes at high levels and in absence of lncRNA UCA1. This research would serve as a first step in understanding and finding better treatment methods for neuroblastoma patients who are resistant to cisplatin drug treatments. As a next step, the validation of UCA1 as a biomarker for drug resistance could serve as novel drug targets and could ultimately lead to the development of antagonists and/or mimics for adjunct therapy with traditional cisplatin treatment methods (Ayers and Vandesompele, 2017). In addition, RNA directed therapy could be used in patients who are highly resistant to the treatment, reducing the discomfort of patients by permitting a lower treatment dosage (Ayers and Vandesompele, 2017). Lastly, a UCA1 biomarker could be quantified in patients through RT-qPCR assays and provide pre-emptive knowledge to oncologists on the best drug combination treatment for each patient (Ayers and Vandesompele, 2017).

Hypothesis

Several studies investigating the molecular mechanisms of lncRNA UCA1 in promoting cisplatin resistance have found that it is involved in silencing of tumor suppressor genes and inducing expression of multidrug resistant proteins (Wang et. al, 2017). We hypothesize that upregulated expression of lncRNA UCA1 is associated with increased resistance to cisplatin in neuroblastoma cell lines, accompanied by changes in the expression of other genes. This will further determine if the role of lncRNA UCA1 is conserved in neuroblastoma compared to other cancer types that have already been studied.

Experimental Plan

Figure 1: Overview of Experimental Plan

Cisplatin resistance after induced changes in lncRNA UCA1 expression

In order to assess whether upregulated expression of lncRNA UCA1 is associated with resistance to cisplatin in neuroblastoma, we will manipulate the levels of UCA1 via knockdown and overexpression in neuroblastoma cell lines. A cell viability assessment will be performed on susceptible SK-N-AS cell line that will be overexpressed with UCA1 and resistant SK-N-ASrCDDP500 cell line that will knockdown expression of UCA1.

Other experiments investigating the function of lncRNA have mapped lncRNA binding sites to the genome after manipulating its expression level (Luo, 2016). However, this technique only gives information on how lncRNA regulates gene expression in cis. Alternatively, the system we propose that manipulates UCA1 levels, will allow us to study the changes in resistance to cisplatin by UCA1, and identify the genes that are potentially affected by UCA1 expression that may lead to resistance. The challenge to our experiment is that when we overexpress lncRNA UCA1 via UCA1 gene transfection, isoforms of the UCA1 gene other than lncRNA UCA1 can be formed. These isoforms may have an effect on cisplatin resistance mechanisms and expression of genes. As a strategy to overcome this challenge, we will silence the isoforms.

As a preliminary experiment, the level of UCA1 expression in cisplatin susceptible SK-N-AS cell line and cisplatin resistant SK-N-AsrCDDP500 cell line will be measured. If lncRNA UCA1 is involved in inducing cisplatin resistance in neuroblastoma, we would expect higher levels of UCA1 in SK-N-AsrCDDP500 cells than in SK-N-AS cells. For the purpose of this project, we design our manipulative experiments assuming that higher levels of UCA1 is observed in the resistant SK-N-ASrCDDP500. If we observe higher UCA1 in susceptible cells, then we should knockdown UCA1 in susceptible cells instead.

Differential Gene Expression Analysis

To quantify changes in RNA expression levels between all conditions, an expression profile analysis will be performed on all RNA samples. RNA will be extracted in equal amounts from all conditions: lncRNA UCA1 knockdown in cisplatin resistant cell lines and lncRNA UCA1 overexpression in cisplatin susceptible cell lines. RNA will also be extracted from susceptible and resistant cell lines with no change in lncRNA UCA1, which serves as a control. All of the extracts will be sequenced and analysed. In comparison to microarrays and qPCR, RNA-seq would allow us to look at differential expression of a broader dynamic range of genes. A summary of the steps and tools that will be used are outlined in Figure 2, with a detailed pipeline in Materials and Methods.

Materials and Methods

Cell lines: Susceptible and Resistant  

Both cell lines will be purchased from the Michaelis Lab, UK. SK-N-AS cells are parental cells of SK-N-ASrCPPD500, which have gained cisplatin resistance. We chose the same lineage of neuroblastoma with the effort to keep the cells to be as genetically similar as possible.

lncRNA UCA1 Overexpression

lncRNA UCA1 will be overexpressed in the susceptible SK-N-AS cell line via plasmid DNA transfection of the UCA1 gene. Both plasmid DNA transfection and RNA transfection are valid methods for overexpression of RNA. However, plasmid DNA transfection is a better method because lncRNA UCA1 will be produced for multiple rounds of replication from the transfected plasmid (Hayashi et al., 2010). In an RNA based transfection, the overexpression of lncRNA UCA1 will rapidly decrease as RNA degrades. A recombinant plasmid of pcDNA-UCA1 will be constructed by inserting UCA1 gene into pcDNA3.1. The recombinant plasmid will be transfected into SK-N-AS cells using Lipofectamine 2000 from Thermo Fisher Scientific (2018). Transfected cells that stably express UCA1 will be selected by RT-PCR. Cells transfected with pcDNA3.1 will also be tested as a negative control to ensure that the transfection procedure does not alter the expression of UCA1 (Wang et. al, 2014).

lncRNA UCA1 Knockdown

Short hairpin RNA (ShRNA) will be used to knockdown lncRNA UCA1 because it is a powerful silencing technique that imitates the endogenous RNA interference mechanism and allows for long term knockdown (Luo, 2016). The scrambled shRNA control (Si-NC) and siRNA that targets lncRNA UCA1 will be purchased from Thermo Fisher Scientific to show that the shRNA protocol does not alter the expression of UCA1. sh-UCA1 lentivirus will be constructed and infected into SK-N-AS cells, which will then be screened with puromycin over 7 days (Fang et. al, 2017). Successful knockdown of lncRNA UCA1 will be assessed using RT-PCR. To prevent UCA1 isoforms from affecting our results, shRNA against isoform RNAs will be transfected as well.

Cisplatin treatment and cell viability assessment  

The cisplatin treatment procedure outlined by Piskareva (2015) will be adapted.  SK-N-AS cells and SK-N-ASrCDDP500 cells will be seeded at 10^4 cells/mL on two separate 96-well plates with 100µL medium per well. The plate will be incubated overnight and will be treated with cisplatin at multiple concentrations the following day. Cell proliferation will be monitored over 5 days. In order to assess the degree to which the cells gain cisplatin resistance, we will perform the MTT assay as described by Wang et al (2008) and therefore measure cell viability.

RNA Extraction

RNA will be extracted from each of the conditions with TRIzol reagent, with twelve replicates for each condition, as recommended by Mortazavi, et al (2008). The purity of total RNA will be evaluated using the A260/A280 ratio of sample absorbance at 260 and 280 nm using NanoDrop ND-1000 (Thermo Fisher Scientific, 2011). Integrity of RNA samples will be measured using the 28S/18S ratio based on a densitometry plot using Agilent 2100 Bioanalyzer.

RNA Illumina Sequencing

RNA-seq will be performed using Illumina HiSeq™ 2000 Sequencing System with paired end sequences for improved accuracy. In order to have a high enough coverage, we will generate ~15-25 million reads per sample, as recommended by Mortazavi, et al (2008). Sequence read quality will be analyzed with the pass/fail metrics of the program FASTQC.

Sequence Alignment

Sequence alignment will be performed using the program STAR and quality of alignment will be measured using RSeQC.

Gene-based Read Counting

Transcript quantification will be calculated using the R package, featureCounts, to generate integer-based read counts for each gene.

Differential Gene Expression Analysis

To analyze the differential gene expression between samples, we will use the R package, DESeq. Genes with a fold change > 2 and p-value < 0.05 will be considered to have a significant change in expression levels. To validate mRNA-seq data, 5 samples will be randomly chosen to run through qRT-PCR for analysis.

Predictions

We predict that knocking down lncRNA UCA1 in SK-N-ASrCDDP500 resistant cell line will result in a rapid decrease in percent cell viability over increasing concentrations of cisplatin and the overexpression of lncRNA UCA1 in SK-N-AS cell lines will result in an increase in percent cell viability.

 

Type of Experiment SK-N-ASrCDDP500 cells (resistant) SK-N-AS cells (susceptible)
lncRNA UCA1 KD % viability rapid decrease
lncRNA UCA1 OX % viability higher than control susceptible cells
Control (no manipulation) % viability higher than susceptible cells Extremely low to zero % viability

Figure 3. Change in cisplatin resistance by knockdown or overexpression of lncRNA UCA1

Differential Gene Expression Analysis

Experiments performing gene expression analysis after manipulating levels of lncRNA UCA1 in the presence and absence of cisplatin continue to show similar changes in RNA expression profiles across cancers that have been studied (Wang et al, 2017). In neuroblastoma, we predict expression analysis to follow suit with other cancers and show a decrease in expression levels in Wnt6, CYP1A1, and AURKC and an increase in expression levels in MBD3 and SRPK1 in the cisplatin resistant knockout lncRNA UCA1 cell line compared to its resistant cell line control (Figure 5). We predict the opposite pattern in UCA1 overexpressed cisplatin susceptible cell line compared to its control, as shown in figure 5.

 

Results

Result 1: Hypothesis is not rejected and predictions are confirmed

We would see a higher percent cell viability when UCA1 is knocked down in the resistant cell line as concentration of cisplatin increases compared to its control, and a higher percent cell viability when UCA1 is overexpressed in the susceptible cell line compared to its control (Figure 4). We would also see changes in expression levels of RNA from the expression analysis in both the knockdown and overexpression of lncRNA UCA1 relative to controls of their respective cell lines (Figure 5). The expression profile of resistant cell line control and susceptible cell line with overexpression would be similar. We can conclude that high levels of lncRNA UCA1 is both sufficient and necessary to increase the percent viability of neuroblastoma cells with increasing doses of cisplatin and to change the expression levels of RNA in the cell lines studied.

 

SK-N-ASrCDDP500 (resistant)              SK-N-AS (susceptible)

 

Figure 4: Percent cell viability of neuroblastoma cells measured in increasing concentration of cisplatin. Percent viability is higher in control SK-N-ASrCDDP500 than knockdown of UCA1. Percent viability is higher in SK-N-AS overexpressed with UCA1 than the control SK-N-AS.

Type of Experiment Expression levels compared to resistant SK-N-ASrCDDP500 control Expression levels compared to susceptible SK-N-AS control
lncRNA UCA1 Knocked down in SK-N-ASrCDDP500 (resistant) Change in RNA expression

For instance:

Downregulated:

Wnt6, CYP1A1, AURKC

Upregulated:

MBD3, SRPK1

may have similar RNA expression patterns
lncRNA UCA1 Overexpressed in SK-N-AS (susceptible) Similar RNA expression Change in expression

For instance:

Upregulated:

Wnt6, CYP1A1, AURKC

Downregulated:

MBD3, SRPK1

Figure 5: Table of possible result 1 for the expression profile analysis

Remarks

We may infer that lncRNA UCA1 regulates a molecular mechanism that induces cisplatin resistance such as silencing of a tumor suppressor gene or inducing expression of multidrug resistant proteins (Wang et al., 2017). Further, if we observe similar changes in RNA expression profiles of neuroblastoma to those in other cancers, we could infer that lncRNA is involved in cisplatin resistance in neuroblastoma in the same pathway found in other cancers.  For example, if we observe a similar upregulation of Wnt6 mRNA in neuroblastoma to that of lung cancer, we may infer that lncRNA UCA1 is involved in cisplatin resistance in neuroblastoma by interactions with Wnt6 in the Wnt pathway (Wang et al., 2008). This is under the assumption that the genes that change in expression are involved in cisplatin resistance rather than unrelated cellular functions. For further investigation into the function of UCA1 and the pathway it is involved in, we can use RNA-TRAP to assess the degree to which lncRNA UCA1 interacts with specific genes and postulate its role in regulating expression of various genes in cis. In addition, we can identify the proteins that lncRNA UCA1 interacts with via crosslinking protein to lncRNA followed by mass spectrometry.

Result 2: Prediction confirmed for overexpression of lncRNA UCA1 in susceptible neuroblastoma cells

We would observe no change in percent cell viability when UCA1 is knocked down in SK-N-ASrCDDP500 compared to its control, but an increase in percent cell viability when UCA1 is overexpressed in SK-N-AS compared to its control. In differential gene expression analysis, where the predictions are satisfied in the cases where genes are overexpressed but not in the knockdown, we would expect to see changes in expression outlined in Figure 7.  We can conclude that high expression of lncRNA UCA1 is sufficient to increase percent cell viability and change the expression levels of RNA of cisplatin susceptible SK-N-AS, but not necessary to maintain percent cell viability and to change the expression levels of RNA in an already resistant SK-ASrCDDP500.

 

SK-N-ASrCDDP500 (resistant)              SK-N-AS (susceptible)

 

Figure 6: percent cell viability of neuroblastoma cells measured in increasing concentration of cisplatin.

 

Type of Experiment Expression levels compared to resistant SK-N-ASrCDDP500 control Expression levels compared to susceptible SK-N-AS control
lncRNA UCA1 Knocked down in SK-N-ASrCDDP500 (resistant) Similar RNA expression may have similar RNA expression
lncRNA UCA1 Overexpressed in SK-N-AS (susceptible) Similar RNA expression Change in expression

For instance:

Upregulated:

Wnt6, CYP1A1, AURKC

Downregulated:

MBD3, SRPK1

Figure 7:  Table of possible results for the expression profile analysis of the second potential result.

Remarks

This result supports the hypothesis that upregulated levels of lncRNA UCA1 may be associated with increased cisplatin resistance, but RNA therapy against only UCA1 would not be sufficient to treat neuroblastoma.  We infer that lncRNA UCA1’s function is redundant to other lncRNAs that induce cisplatin resistance. This is assuming other lncRNAs could have similar to identical functions in regulating gene expression and that the expression of these other lncRNAs are high and saturated in the control resistant SK-N-ASrCPPD500 cells. For example, the alteration of the wnt pathway is a common way of inducing cisplatin resistance and several lncRNA such as HOTTIP and MALAT have been identified to affect this pathway (Hu et. al, 2018).  As a next step, we could compare the expression profiles resulting from manipulation of putative lncRNAs with that of lncRNA UCA1. In addition, we could study the proteins that lncRNA UCA1 interacts with via CHART technique and compare with other lncRNAs.

Result 3: Hypothesis rejected with no change in cell viability and expression profile

In the case where there is no change in percent cell viability and RNA expression profile after manipulation of UCA1 levels, our hypothesis will be rejected. We conclude that lncRNA UCA1 is neither sufficient nor necessary to increase percent cell viability and change the RNA expression profile in the neuroblastoma cell lines of study treated under multiple concentrations of cisplatin. We cannot conclude that lncRNA UCA1 has no function in neuroblastoma, because it is still possible that it is involved in other ways.

 

SK-N-ASrCDDP500 (resistant)              SK-N-AS (susceptible)

Figure 8: percent cell viability of neuroblastoma cells measured in increasing concentration of cisplatin.

 

Type of Experiment Expression levels compared to resistant SK-N-ASrCDDP500 control Expression levels compared to susceptible SK-N-AS control
lncRNA UCA1 Knocked down in SK-N-ASrCDDP500 (resistant) Similar RNA expression Different expression

  • This will be the same differences in expression that were found when comparing the resistant and susceptible controls
lncRNA UCA1 Overexpressed in SK-N-AS (susceptible) Different expression

  • This will be the same differences in expression that were found when comparing the resistant and susceptible controls
Similar RNA expression

Figure 9: Table of results if both the hypothesis and prediction are rejected.

 

Assuming that lncRNA UCA1 has all of the requirements to perform its normal functions in the in vitro experiment, we can infer that lncRNA UCA1 does not have a function in the pathways that induce or reduce cisplatin resistance in our neuroblastoma cell lines. Previous studies have found other lncRNAs apart from UCA1 can induce cisplatin resistance, so it is possible that the UCA1 knockdown resistant cell line is resistant to cisplatin because of other lncRNAs and proteins. To investigate these, RNAs expressed differently in SK-N-ASrCDDP500 control compared to SK-N-AS control could be investigated using a similar method to this experiment. Once confirmed of their potential involvement in cisplatin resistance, we can further study their function by assessing the proteins they interact with and the genes that they regulate expression of, as described in result 1 and 2. Additionally, if we observed higher levels of UCA1 in the resistant cell line control compared to the susceptible control, we might infer that upregulated UCA1 is a consequence of cisplatin resistance rather than the cause.

Citations

 

Ayers, D., & Vandesompele, J. (2017). Influence of microRNAs and long non-coding RNAs in cancer chemoresistance. Genes, 8(3), 95. doi:10.3390/genes8030095

 

Balas, M. M., & Johnson, A. M. (2018). Exploring the mechanisms behind long noncoding RNAs and cancer. Non-Coding RNA Research, 3(3), 108-117. doi:10.1016/j.ncrna.2018.03.001

 

Chen, Q., Wei, C., Wang, Z., & Sun, M. (2017). Long non-coding RNAs in anti-cancer drug resistance. Oncotarget, 8(1), 1925-1936. doi:10.18632/oncotarget.12461

 

Dasari, S., & Tchounwou, P. B. (2014). Cisplatin in cancer therapy: molecular mechanisms of action. European journal of pharmacology, 740, 364-78.

 

Eastlack, S. C., Dong, S., Mo, Y. Y., & Alahari, S. K. (2018). Expression of long noncoding RNA MALAT1 correlates with increased levels of nischarin and inhibits oncogenic cell functions in breast cancer. PloS One, 13(6), e0198945. doi:10.1371/journal.pone.0198945

 

Fan, Y., Shen, B., Tan, M., Mu, X., Qin, Y., Zhang, F., & Liu, Y. (2014). Long non-coding RNA UCA1 increases chemoresistance of bladder cancer cells by regulating Wnt signaling. FEBS Journal,281(7), 1750-1758. doi:10.1111/febs.12737

 

Galluzzi, L., Senovilla, L., Vitale, I., Michels, J., Martins, I., Kepp, O., . . . Kroemer, G. (2012). Molecular mechanisms of cisplatin resistance. Oncogene, 31(15), 1869-1883. doi:10.1038/onc.2011.384

 

Guttman M, Donaghey J, Carey BW, Garber M, Grenier JK, Munson G, Young G, Lucas AB, Ach R, Bruhn L, Yang X, Amit I, Meissner A, et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature. 2011; 477:295–300.

 

Hayashi, T., Lamba, D., Slowik, A., Reh, T., & Bermingham-McDonogh, O. (2010). A method for stabilizing RNA for transfection that allows control of expression duration. Developmental Dynamics, 239(7), 2034-2040. doi:10.1002/dvdy.22344

Hu, Y., Zhu, Q., Deng, J., Li, Z., Wang, G., & Zhu, Y. (2018). Emerging role of long non-coding RNAs in cisplatin resistance. OncoTargets and Therapy, 11, 3185-3194. doi:10.2147/OTT.S158104

 

Luo, M. (2016). Methods to study long noncoding RNA biology in cancer. (pp. 69-107). SINGAPORE: SPRINGER-VERLAG SINGAPORE PTE LTD. doi:10.1007/978-981-10-1498-7_3

 

Malik, R., Patel, L., Prensner, J., Shi, Y., Iyer, M., Subramaniyan, S., . . . Chinnaiyan, A. (2014). The lncRNA PCAT29 inhibits oncogenic phenotypes in prostate cancer. Molecular Cancer Research, 12(8), 1081-1087. doi:10.1158/1541-7786.MCR-14-0257

 

Moran VA, Perera RJ, Khalil AM. Emerging functional and mechanistic paradigms of mammalian long non-coding RNAs. Nucleic Acids Res. 2012; 40:6391–6400.

 

Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B (2008) Mapping and quantifying mammalian transcriptomes by RNAseq. Nat Methods 5:621–628.

 

Piskareva, O., Harvey, H., Nolan, J., Conlon, R., Alcock, L., Buckley, P., . . . Stallings, R. L. (2015). Corrigendum to “The development of cisplatin resistance in neuroblastoma is accompanied by epithelial to mesenchymal transition in vitro” [Cancer Lett 364 (2015) 142–155]. Cancer Letters,369(2), 428. doi:10.1016/j.canlet.2015.09.010

 

Ponting, C. P., Oliver, P. L., & Reik, W. (2009). Evolution and functions of long noncoding RNAs. Cell, 136(4), 629-641. doi:10.1016/j.cell.2009.02.006

 

Thermo Fisher Scientific. (2011). NanoDrop 1000 Spectrophotometer V3.8 User’s Manual. Retrieved from: http://tools.thermofisher.com/content/sfs/manuals/nd-1000-v3.8-users-manual-8%205×11.pdf

 

Wang, H., Guan, Z., He, K., Qian, J., Cao, J., & Teng, L. (2017). LncRNA UCA1 in anti-cancer drug resistance. Oncotarget, 8(38), 64638-64650. doi:10.18632/oncotarget.18344

 

Wang F, Li X, Xie X, Zhao L & Chen W(2008) UCA1, a non‐protein‐coding RNA up‐regulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion. FEBS Lett 582, 1919–1927.

 

Wang, Q., Armenia, J., Zhang, C., Penson, A. V., Reznik, E., Zhang, L., . . . Schultz, N. (2018). Unifying cancer and normal RNA sequencing data from different sources. Scientific Data,5, 180061. doi:10.1038/sdata.2018.61

 

Wang, Y., Zhang, D., Wu, K., Zhao, Q., Nie, Y., & Fan, D. (2014). Long noncoding RNA MRUL promotes ABCB1 expression in multidrug-resistant gastric cancer cell sublines.Molecular and Cellular Biology, 34(17), 3182-3193. doi:10.1128/MCB.01580-13

 

Wang, F., Zhou, J., Xie, X., Hu, J., Chen, L., Hu, Q., . . . Yu, C. (2015). Involvement of SRPK1 in cisplatin resistance related to long non-coding RNA UCA1 in human ovarian cancer cells. Neoplasma,62(03), 432-438. doi:10.4149/neo_2015_051

Wang, F., Li, X., Xie, X., Zhao, L., & Chen, W. (2008). UCA1, a non-protein-coding RNA up-regulated in bladder carcinoma and embryo, influencing cell growth and promoting invasion. FEBS Letters,582(13), 1919-1927. doi:10.1016/j.febslet.2008.05.012

 

Wang, B., Huang, Z., Gao, R., Zeng, Z., Yang, W., Sun, Y., . . . Zhou, S. (2017). Expression of Long Noncoding RNA Urothelial Cancer Associated 1 Promotes Cisplatin Resistance in Cervical Cancer. Cancer Biotherapy and Radiopharmaceuticals,32(3), 101-110. doi:10.1089/cbr.2016.2156

 

Zhou, X., Liu, S., Cai, G., Kong, L., Zhang, T., Ren, Y., . . . Wang, X. (2015). Long non coding RNA MALAT1 promotes tumor growth and metastasis by inducing epithelial-mesenchymal transition in oral squamous cell carcinoma. Scientific Reports, 5(1), 15972. doi:10.1038/srep15972

 

Final updated honeybee model

After creating the honeybee model, we received feedback from Pam and my response:

  •  The involvement of TOR is not clear in our model
    • TOR gene regulates YAP, a component of the HIPPO pathway
  • How could royalactin regulate the EGFR pathway
    • the royal actin acts as a ligand and binds to receptors in of the EGFR pathway thereby inducing the pathway.
  • Where does DNMT3 come into play
    • DNMT3 is activated by p-cumaric acid in bread is involved in promoting the worker phenotype
  • How could 10HDA affect physio-metabolic gene’s expression?
    • maybe inhibiting the action of deacetylases?

Figure 1. model of factors that regulate honeybee caste differentiation with a focus on the EGFR pathway.

Group members: Joanne, Melissa, Meilin, Julie-Anne, Evan Z, Tanis

Summary of the model: The egfr pathway is activated by p-cumaric acid and royal actin. egfr regulates MAPK and S6K that affects phenotype of honeybees. 10HDA and p-cumaric acid may be involved in differential gene expression, leading to specific castes.

Gaps in evidence: We think that 10HDA may affect the physio-metabolic genes, but there is no evidence to support this statement. The link between 10HDA and egfr pathway is unclear.

A suggestion for the two most critical or most interesting experiments that the community should next focus on: Study the role of 10HDA in the egfr pathway and on the physio-metabolic genes. Can do a knockdown and overexpression experiment and see the changes of expression profile of genes in the egfr pathway or physiometabolic genes

REFERENCES:

Ashby, R., Forêt, S., Searle, I., & Maleszka, R. (2016). MicroRNAs in honey bee caste determination Scientific Reports, 6(1), 18794. doi:10.1038/srep1879

Kamakura, M. (2011). Royalactin induces queen differentiation in honeybees. Nature, 473(7348), 478-483. doi:10.1038/nature10093

Li, J., Issa, J. J., Bedford, M. T., Castellano, S., Raynal, N. J. -., Gharibyan, V., . . . Spannhoff, A. (2011). Histone deacetylase inhibitor activity in royal jelly might facilitate caste switching in bees. EMBO Reports, 12(3), 238-243. doi:10.1038/embor.2011.9

Kucharski, R., Foret, S., & Maleska, R. (2015). EGFR gene methylation is not involved in royalactin controlled phenotypic polymorphism in honey bees. Scientific Reports, 5(1), 14070. doi:10.1038

Mao, W., Schuler, M. A., & Berenbaum, M. R. (2015). A dietary phytochemical alters caste-associated gene expression in honey bees. Science Advances, 1(7) doi:10.1126/sciadv.1500795

Zhu, K., Liu, M., Fu, Z., Zhou, Z., Kong, Y., Liang, H., . . . Chen, X. (2017). Plant microRNAs in larval food regulate honeybee caste development. PLoS Genetics, 13(8), e1006946. doi:10.1371/journal.pgen.1006946

 

Top4: Quiz 4

Joanne (5)

My quiz was on the Hillman et al. paper. I attached my quiz results above. It was an open book quiz asking us to interpret data and give our own opinion about the data and the author’s conclusions. They studied the expression of UBE3A in the CNS and non CNS to see if there is genomic imprinting that regulates the dosage of UBE3A in neurons. I chose this as my top 4 assignment because it was my first time coming up with my own opinion that would go against the authors’ conclusion. The authors had the assumption that imprinting results in a reduced dosage which lead to the conclusion that imprinting doesn’t regulate dosage of UBE3A. However, UBE3A is clearly higher in the CNS than non CNS, so if we think of this case in another way, apart from their assumption, genomic imprinting may be associated with higher level of UBE3A in the CNS.

Below is the worksheet that my friends and I worked on in preparation for the quiz. [there was misformatting when transferring the document such that the numbers are off]

Figure 1: expression of UBE3A in mouse and human CNS (eutherian)

  1. What is FPKM normalization?
    • This stands for fragments per kilobase million from the RNA seq data and it’s a a way of reporting RNA seq results. Normalized here just means that it is per million bases so is is a normalized number rather than being per some other number that is different between RNA seq data (want all data to be reported using same units)
    • Why is there no significance of ube3a in hippocampus in FPKM normalization?
      • There is a significant difference between all samples but just not specifically between hippocampus compared to liver and thymus (but there is a sig diff between hippocampus and heart and lung)
  2. What question is addressed in the experiment?

To test if imprinting in neurons (in the paternal allele) of UBE3A reduces dosage of UBE3A in the neurons compared to other cell types  

  1. Why did the authors have a Ube3am+/p-?
    • To estimate the relative expression levels of the maternal Ube3a allele (to control for the potential effects of the paternal allele on the expression levels)
  2. Justify their choices in technique, control.. Etc.
  3. What could the authors have done for better results?
  4. What do the data show?
  • Higher Ube3a transcript level in the CNS compared to the N-CNS in both Ube3a m+/p+ and Ube3aM+/p-
  • a) Ube3a has significantly higher transcript levels in the CNS (cortex, hippocampus) compared to other cells (heart, kidney, etc) in both Ube3a m+/p+ and Ube3aM+/p-
  • b) Ube3a has significantly higher protein levels in the CNS (cortex, hippocampus) compared to other cells (heart, kidney, etc) in both Ube3a m+/p+ and Ube3aM+/p-
  • c) There is no significant difference between the CNS of Ube3a m+/p+ and Ube3aM+/p- in both the protein and transcript expression
  1. What can we directly conclude from the data?
    • Functional WT paternal Ube3a allele is not necessary to have WT levels of Ube3a transcript in both the CNS and N-CNS tissues (fig 1a)
    • Though hippocampus tissues have a large 5 fold difference between maternal allele and paternal allele transcript abundance, its overall Ube3a transcript count is comparable to other tissues, like the thymus, heart, and liver.
  2. In one sentence, what did the authors demonstrate?
    • CNS transcript abundance of Ube3a in human and mice are similar to many Non-CNS tissues and the maternal Ube3a allele produces the bulk of the transcript product in  CNS.
  3. Are these results what you would expect? Why?
    • In some ways yes and in some no. I would expect to see equal or lower expression levels of Ube3a in CNS compared to N-CNS since in the CNS, Ube3a is imprinted and therefore there is only expression coming from the maternal allele, however, we observe that Ube3a is expressed in higher amounts in the CNS compared to N-CNS. This suggests that Ube3a is being overexpressed in the maternal allele or there is some sort of regulatory mechanism happening when the paternal allele is expressed. Other results are expected.

 

Figure 2: Maternal Ube3a compensates for loss of paternal Ube3a expression during neurogenesis

  1. What experiments did they do? What are the controls and why did they have that?

a)Immunofluorescence imaging of prenatal primary neurospheres

Maybe no control:  they compared the pattern in paternal and maternal Ube3a

  1. b) western blot to show the difference in YFP and normal protein
  2. d) measured intensity values  
  1. What was the purpose of these experiments? What question were they addressing?
  • This was to show that imprint is acquired during neuronal differentiation and there is bias toward maternal allelic expression of Ube3a in the days post differentiation
  • Question addressed: does the maternal allele shift expression in a compensatory way for the paternal decreased expression?
  1. What do the data show?
  1. d) intensity of maternal UBe3a increases significantly at 16DPD. Paternal decreases in a linear manner from DPD1 to DPD16. There seems to be an inverse relationship between the maternal and paternal Ube3a in the primary neurons
  2. e) At DPD1, the difference in intensity of Ube3a is not significant. As DPD increases, the discrepancy between maternal and paternal protein intensity increases.
  1. What can we conclude from the data?

There seems to be an inverse relationship between the maternal and paternal Ube3a in the primary neurons. Intensity of maternal protein increases while that of paternal protein decreases.  

  1. What can we not conclude?

Whether or not the decrease in paternal protein intensity CAUSES increase in maternal intensity

  1. What can we infer from the data?

Imprinting of ube3a at the paternal allele is initiated upon neuronal differentiation. Increase in Ube3a from the maternal allele compensates for loss of expression of the paternal allele in neurons.

Figure 3: Ube3a protein levels are higher in mouse and opossum despite no imprinting in opossum

 

  1. What does the several SNV mean?
  1. Single nucleotide variant
  1. What experiments did they do? What are the controls and why did they have that?

 

  1. What was the purpose of these experiments? What question were they addressing?
  2. What do the data show?
  1. The alleleic ratio of maternal and paternal in UBE3A (opossum) are similar in the SNVs of the cortex. However, the number for paternal is slightly higher in SNV4 than maternal though not statistically significant.
  2. Expression of UBE3A in CNS is significantly higher than in N-CNS
  1. What can we conclude from the data?
  1. UBE3A is biallelically expressed in the opossum
  2. Imprinting is NOT NECESSARY to result in high UBE3A expression in CNS compared to N-CNS
  1. What can we not conclude?
  1. What can we say about slightly higher ratio in paternal?
  2. If imprinting is not the cause for higher UBE3a expression in imprinted mouse  
  1. What can we infer from the data?
  1. b) There are other mechanisms that induce expression of mouse UBE3A particularly in the CNS compared to N-CNS
  1.     C) imprinting in the mouse may cause higher expression of Ube3a in the cortex compared to expression of UBE3A in opossum.

Top4: wang et al.

This paper was enjoyable to read and gave us space to design a model from their findings. Although the model may not have been good, it was a great practice to combine small pieces of information to figure out the big picture. My group focused on Question 4 – that lncRNA HOTTIP associates with CTCF protein. This was also good background information for my group’s final project when talking about future directions to study the function of a lncRNA.

Small cooperative assignment: Wang et al. paper

Assigned paper: Wang, F., Tang, Z., Shao, H., Guo, J., Tan, T., Dong, Y., Lin, L. (2018). Long noncoding RNA HOTTIP cooperates with CCCTC-binding factor to coordinate HOXA gene expression. Biochemical and Biophysical Research Communications 500 (2018) 852e859

A note about HOTAIR (a very famous line RNA involved in hox regulation): We will see in detail how another lncRNA, air, carries out the same function, but at an imprinted locus. So, don’t worry too much about it twhen it is discussed in the introduction.

Question 1 (everyone)

Before this paper, what was known to the authors about HOTTIP, what it does, and how it drives methylation of H3K4? What is the consequence of having a lot of H3K4me at a locus?

HOTTIP is a lincRNA, found in the HOXA locus.

Question 2 (Erin, Melissa, Elizabeth, Miguel, Cathy, Oline, Jasia)

Consider Figure 1.

  1. What was the experiment that lead to those results? (What did the authors do, what did they measure)?
    • Used published data of:
      • ChIP-seq with CTCF, H3K4me3, and H3K27me3 at the HoxA locus
      • RNA-seq of HoxA (A1 – A13)
  1. What question were the authors addressing in the experiment?
  • What role does CTCF have on transcriptional control in Hox genes through a model of chromatin structure organization?
  • Does the amount of histone modification (H3K27me3 and H3K4me3) correlate to RNA transcripts of the HoxA genes?
    • (edited from: the location of the HoxA genes? )
  • What is the spatial relationship between CTCF binding sites to HoxA genes?
  1. Why did the authors (most likely) chose this two particular cell lines?
  • BJ and HFF cells are distal foreskin types of fibroblast cell lines and expression of HOTTIP was mainly observed in human fibroblasts and are not fully differentiated yet.
  • They are distal, being from foreskin and hoxA genes are expected to be expressed in distal stages
  1. In terms of the ChIP experiments, what additional control could the authors have included, and what would its purpose have been? [If you have trouble with this question, set it aside and come back to it at the end].
  • Measured the amount of histone modifications in other cell lines, not fibroblast lines, as a negative control
  • Measured the amount of different types of histone modifications in HoxA genes to show that specific histone modifications are involved in the expression of the HoxA genes
  • For C and D, they can also test the CTCF antibody specificity at other DNA sites (other than the C1-C6 regions) that we believe do not associate with CTCF
  1. What do the data show?
  • Hox A transcripts are present at higher levels from regions that are also enriched for H3K4me3, which is typically associated with transcriptionally active regions
  • CTCF is associated in  regions C1-6
  1. What can we directly conclude from the data, and why?
  • There is a direct relationship between H3K4me3 levels and levels of HoxA RNA transcript
  • There is an inverse relationship between the H3K27me3 levels and levels of HoxA RNA transcript
  1. In one sentence, what did the authors demonstrate?
  • The authors conducted an observation experiment to lay out groundwork on putative CTCF binding sites and demonstrated a correlation between histone modifications enrichment and expression of hoxa cluster genes.

Question 3 (Tanis, Shannah, Meilin, Pareesa, Dee, Maria S, Amanda)

Consider Figure 2.

  1. What were the experiments that lead to those results? (What did the authors do, what did they measure)?  

Knock out C5 (CRISPR) on HOXA gene cluster. They measured CTCF binding to C1-C6 domains on HOXA gene in BJ cells/HFF cell lines using ChIP assays. They also measured mRNA expression of HOXA genes when they knocked out C5 and C5 and HOTTIP knockout (RNAI with siRNA) and compared them to a wild type control.

 

  • What questions were the authors addressing in the experiments?

 

Is there a functional connection between HOTTIP lincRNA and CTCF binding factors? If there is, how does it contribute to gene activation and chromatin organization? Do we observe effects on HOXA gene expression when we disrupt CTCF binding (via C5 region deletion on HoxA)  and knockdown HOTTIP respectively (via RNAi)?

Panel C: Is there differential HOTTIP RNA expression between BJ and HFF cells?/ Is there HOTTIP expression in living cells?

 

 

  • The authors did a HOTTIP knock-down (siRNA) and not a knockout of the HOTTIP locus. Please provide two reasons why a knockout would not have been appropriate.

 

  1. Knock-down siRNA doesn’t disrupt the HOXA gene, as only lincRNA is affected. If they did a knockout by removing HOTTIP, it would not be clear whether the observed effect was due to the knockdown itself or the disruption of the HOXA gene itself.
  2. SiRNAs are extremely specific. The nucleotide sequence will specifically match the HOTTIP lincRNA. In comparison a knockout is much less specific, which could potentially lead to unintentional effects.

 

  • What is the purpose of running the experiments on two different cell lines?

 

To ensure that the observed effects are not specific to the cell line.

 

  • What do the data show?

 

A+C – Deletion of CTCF core motif leads to abrogation of CTCF occupancy at C5 site in both BJ and HFF cells. Sites C4 and C6 are also affected and there is a large decrease in CTCF binding.

B+D – Deletion of C5 site leads to upregulation of HOXA6 (gene immediately rostral of C4 and C5 sites) in comparison to WT control. HOXA3-5 have increased expression whereas HOXA1 and HOXA2 are fully repressed.

When HOTTIP was knocked down, this abrogated the upregulation effect of HOXA3-6 genes after C5 deletion.

 

  • Are any of the data surprising? If so, which parts?

 

There was a dramatic decrease in CTCF binding at the neighbouring sites C4 and C6.

 

  • What can we directly conclude from the data, and why?

 

We can directly conclude:

  • C5 is necessary to bind CTCF to HOXA gene clusters, at C5 and also is necessary for maximal C4 and C6 to CTCF.
  1. In one sentence, what did the authors demonstrate?

CTCF and HOTTIP coordinate together to sho

 

Question 4 (Heather, Ana-Maria, Brett, Jamie, Jacob, Josh, Beth)

Consider Figure 3.

  1. What was the experiment that lead to those results? (What did the authors do, what did they measure)?
    1. The experiment used a CRISPR-mediated gene KO of the ∆C5 CTCF binding element within the HoxA DNA cluster. Histone methylation marks H3K27me3 and H3K4me3 were measured using ChIP-Seq against two different human foreskin fibroblast cell lines, BJ and HFF. By this method, the authors were able to determine the presence and quantity of the two histone methylation marks at the various HoxA loci, A1-A13. As a final manipulation, the authors also used an siRNA-mediated knockdown of the HOTTIP lncRNA, followed by ChIP-seq, to study the effects of HOTTIP on HoxA locus histone methylation
  2. What question were the authors addressing in the experiment?
    1. The authors wanted to investigate how HOTTIP and CTCF are related to HoxA gene expression levels through chromatin modifications (methylation of histone subunits).
  3. What additional ChIP control could the authors have included, and what would its purpose have been? [If you have trouble with this question, set it aside and come back to it at the end].
  4. The authors did a HOTTIP knock-down (siRNA) and not a knockout of the HOTTIP locus. Please provide two reasons why a knockout would not have been appropriate.
    1. Because Hox genes are expressed in a collinear manner; i.e. they are expressed in a particular order along the body axis during development, a knockout of the HOTTIP locus may result in disruption of the spatial regulation and the HoxA cluster, whereas siRNA would have no effect on the spatial location of the locus.  
    2. In addition, an RNA knock-down allows temporal control of the experiment, so that the siRNA can be reversed if necessary
    3. Ensures no disruption of the hox cluster structurally or sequence wise
  5. What do the data show?
    1. In BJ cells, the H3K4me3 mark across loci A13-A7 were almost identical (capturing ~80% of the input) in terms of presence and quantity between WT and ∆C5 cells; siHOTTIP + ∆C5 cells had between 20 and 40% of the total input capture. Significantly reduced H3K4me3 was seen in WT cells across the A6-A2 locus relative to ∆C5 cells (WT cells captured between 5-20% of the input), and even presence/quantity of the mark across all conditions at the A1 locus.
    2. Also in BJ cells, the H3K27me3 mark presence across loci A1, A2, and A7-13 between the WT, ∆C5 and ∆C5+siHOTTIP cells were fairly identical, though the percentage of input captured varied, with there being a fairly constant gradient from the lowest input covered in A13-10 (~8%) to the highest input covered (~50%) in A1. However, the WT percent capture in A3-6 was on average 2 fold greater than that of the ∆C5 and ∆C5+siHOTTIP knockdowns, hovering around 50% whereas the ∆C5 and ∆C5+siHOTTIP knockdowns decreased over the A3-A6 gradient, from ~30% in A3 to ~20% in A6. This indicates significantly reduced H3K27me3 in those loci in the knockdown experimental groups.
    3. In HFF cells, there is no significant difference in
  6. Are any of the data surprising? If so, which parts?
    1. Functional lincRNAs are not well characterised, and so although they may be abundant, it is still a surprising/rare phenomenon to come across in gene regulation experiments at the moment
  7. What can we directly conclude from the data, and why?
    1. The C5 binding site on the HoxA locus is required for the normal (WT) H3K4me3 (“activating”) and H3K27me3 (“silencing”)  histone modifications levels respective to the HoxA genes. The addition of an siRNA targeting the HOTTIP RNA showed that HOTTIP is necessary for the H3K4me3 histone modifications on the HoxA genes, but not for the H3K27me3 histone modifications in both BJ and HFF cells.
  8. In one sentence, what did the authors demonstrate?
    1. The authors demonstrated that HOTTIP in coordination with CTCF binding  is required for maintenance of epigenetic modifications that regulate expression of the HoxA genes in foreskin fibroblasts.

 

Question 5 (Kate, Maria V., Evan G., Joanne, Evan Z, Hayden)

Consider Figure 4.

 

  • What were the experiments that lead to those results? (What did the authors do, what did they measure)? You may have to separate out the different parts of the figure (different experiments).

 

PANEL A: The authors engineered E. coli cells to express recombinant GST and GST-tagged CTCF. They purified these proteins, and ran them on a gel. Panel A shows this gel, to demonstrate that they successfully purified these proteins.

 

PANEL B: The purified GST and GST-CTCF were incubated with HOTTIP RNA, or control histone mRNA. Any RNA not bound to these purified proteins was washed away. RT-PCR was used to show whether either protein was able to pull down HOTTIP RNA, as compared to the histone control which is known to bind to neither protein.

 

PANEL C: This shows the results of an IP experiment, pulling down the CTCF protein from cultured human foreskin fibroblasts. The authors then use RT-PCR to measure the amount of HOTTIP RNA was found to be associated with their immunoprecipitated CTCF protein, as compared with the negative control IP for IgG.

 

PANEL D: This is an in vivo experiment, wherein the authors use an anti-biotin antibody to pull down biotinylated HOTTIP RNA, along with GFP and antisense HOTTIP RNA to serve as negative controls. They separate any associated proteins, and run then on a gel. They probe for CTCF, WDR5 (a protein to which HOTTIP is known to associate) and a-tubulin (a protein to which HOTTIP is known to have no association).

 

 

  • What question were the authors addressing in each of the experiments?

 

Figure 4A – Have we successfully purified the recombinant proteins that we will be using to test interaction between HOTTIP and CTCF

Figure 4B- To what levels do HOTTIP RNA and GST-CTCF interact in comparison to GST?

Figure 4C- Is there differential HOTTIP RNA expression between BJ and HFF cells?/ Is there HOTTIP expression in living cells?

Figure 4D – Does HOTTIP bind to CTCT? Does HOTTIP retrieve CTCF?

 

 

  • What is the purpose of the histone mRNA in panel B

 

Histone mRNA is a control to show that the induced retrieval of HOTTIP RNA by GST-CTCF compared to GST is due to specific interactions of HOTTIP and GST-CTCF rather than random chance. Histone mRNA is a negative control in which we do not expect induced retrieval in either of the proteins. Because the levels of histone mRNA retrieved by GST and GST-CTCF are the same, we know that the experimental system is valid (?) and describe that HOTTIP RNA retrieval by HOTTIP RNA is higher.

 

 

  • What do the data show?

 

Figure 4A – Western blot shows successful purification of GST-CTCF, which were used to test HOTTIP RNA binding/retrieval in vitro

Figure 4B – GST-CTCF and not GST (control) retrieved HOTTIP RNA and not histone mRNA (control), as measured by qRT-PCR

Figure 4C – HOTTIP RNA is co-precipitated with CTCF but not with IgG IP (control) in both BJ and HFF cells

Figure 4D – HOTTIP RNA, and not antisense HOTTIP RNA or GFP, retrieves CTCF in vivo, and WDR5 (positive control), but does not retrieve alpha-tubulin (negative control)

 

 

  • What can we directly conclude from the data, and why?

 

We can directly conclude that HOTTIP RNA directly interacts with CTCF in vivo and in vitro.

  1. What would be your next experiment, if you were one of the authors?

HOTTIP is a lncRNA with 3764 nucleotides, so I would attempt to identify the region of HOTTIP that interacts with CTCF. To do this I would amplify HOTTIP RNA, shear it into smaller fragments of RNA, use CTCF to pull out RNA from this sample, and sequence the resulting RNA. From there, I could build a consensus sequence from the various sequenced fragments. This consensus sequence would likely indicate the specific region within HOTTIP that interacts with CTCF.

 

  • In one sentence, what did the authors demonstrate?

 

  • Results reveal that there is a functional connection between HOTTIP and CTCF, and sheds light on lincRNAs (long intergenic non coding RNA) involvement in gene activation.

 

life science research night 2018

Sometime in November, I attended the life science research night that was in the life science research institute. I went into an info session about applying to graduate schools as well as an introduction to drug development. At the very end, I wandered around through different posters. It was interesting how my experience was different from when I attended this event in my second year. In my second year, I wasn’t able to follow through most of the posters and was unfamiliar with the terminology that presenters were using. This time, I had a much better understanding of what the presenters are talking about. For example – about integrins, about histone modification, about DNA methylation, about cancer metastasis etc. The one that stood out to me the most was ” Identification of Novel Lung Cancer Drivers from a Sleeping Beauty Mutagenesis Screen”. It was related to identifying cancer-promoting genes and studying the function of those genes. I had the chance to ask the presenter how she came to work on this project. She talked about her co-op experience in the lab and how she decided to continue working on the project in grad school. It was interesting how long a project takes. She said that the project started before she was doing co-op then there was no progress for almost a year (although there was ongoing effort) and then they finally got some results that work to allow them to progress further. –> I think this can be an example of persistence? (kind of related to self-regulated learning)

Pathway to our final final project

I really enjoyed working on this project. We started by reading papers related to lncRNA and cancer. Then after having a discussion with Evan and Pam we found the direction to study the mechanism of lncRNA in chemotherapy resistance. It was fun finding a gap in information/ research results in the academic world (?). Progressively, we narrowed down to cisplatin as our drug and neuroblastoma as our cancer type. Although not included in the final project, I learned so much about techniques related to studying the function of lncRNA and how they are involved in other processes. I think focusing on lncRNA as my project also helped me understand lectures related to lncRNA better.

DocuLearn 5

Questions

  1. Please review your DocuLearn 1 to remind yourself of the two “things” that you wanted to learn in the course. Did you learn them (to the level that you were expecting)? If not in this course, did you learn them in a different course or context this term?

 

      • Through this course, I became more familiar with reading review articles and research papers to gain more knowledge in gene regulation processes. I learned that searching for more articles and skimming the abstract and introduction helps to gain understanding that may not have been provided by one article. My expectations were met.
      • I also learned to ask questions that will help me gain more understanding of the system of study. For example: what do the data show, what can I conclude, what can I infer, can I think of a way to explain the results? My expectations were met for this one as well.

 

2. Recall the idea saying that self-regulated learning involves:

      • attending to key features of the environment
      • resisting distractions
      • persisting when tasks are difficult
      • responding strategically, flexibly adaptively.

Were there situations in the course when you feel that you really demonstrated each of these behaviours (and what were the situations)?

      • Resisting distractions: One of the biggest challenge while working on the final project was spending too much time reading articles that may not be important. It would be great if I could figure that out early on, but for this occasion, it was a good opportunity to get exposed to any related topics.
      • Persisting when tasks are difficult: I think I would say I accomplished this if I successfully hand in my project. Also sought for help when we felt like we were stuck.
      • Responding strategically, flexibly and adaptively: I’m not sure what to say about this point. We have spent so much time on the draft and it’s still not finished. If I have time, I would like to ask my peers of what they thought about this point. What other instances might there be that I am overlooking?
  1. Overall, what was the most challenging thing in the course for you? How did you handle this challenge?
      • Learning from other people’s perspectives and speculations. Initially, I had a hard time following through people’s ideas. I think later on I started to understand better of what classmates are discussing. Sometimes writing down the discussion word for word then looking back at it helped. I think another factor that helped was having a better understanding of what is going on in the system that we are talking about.
      • Another challenge: time management! I can’t say that I overcame this challenge. I think I could have been more active in discussions if I did the reading beforehand. I thought I was pretty fine with time management, but I’m not sure what to say in the context of this course. I think I’ll have a better thought after completing the final project…  I think the problem is that I spend too much time on unnecessary things? Or not? I will get back to this part if I have time.

From <https://canvas.ubc.ca/courses/9305/assignments/261593>