| CJUH-JLU-China - iGEM 2023

Results

Overview

The goal of our project is to develop a breast cancer screening kit, BreFast. Our project integrated the knowledge of computer science, molecular biology and biochemistry. We used bioinformatics approaches to analyze breast cancer-associated miRNA (microRNA), and selected miR-21-5p as the best biomarker for detection by BreFast. Subsequently, we verified the expression of miR-21-5p in breast cancer cells by qRT-PCR, and explored the function of miR-21-5p by wound healing and transwell assays. Furthermore, we designed a crRNA, an essential component of CRISPR/Cas13a system, to target miR-21-5p, and made an engineered CRISPR/Cas13a system that could be activated by miR-21-5p. The cleavage efficiency of the CRISPR/Cas13a system was investigated by fluorescence reaction and kinetics simulation. Finally, the efficacy of the CRISPR/Cas13a system on detecting high levels of miR-21-5p was demonstrated with lateral flow strips. In summary, we have combined CRISPR/Cas13a system and lateral flow strips into a convenient and reliable kit to detect miRNA associated with breast cancer (Figure 1).

Figure 1 The workflow of the Results section.

miR-21-5p

Bioinformatics Analysis

During the progression of breast cancer, the expression levels of miRNAs are changed. Some of these miRNAs are closely associated with breast cancer and can be used as potential biomarkers for detection of breast cancer. We focused on utilizing miRNAs that show increased expression in breast cancer as biomarkers for detection by BreFast.

Differential expression analysis

To identify miRNA with significant changed expression in breast cancer patients, we analyzed TCGA-BRCA dataset in TCGA database and GSE59247 dataset in GEO database, and selected miRNA with thresholds of |logFC|>1 and PValue<0.05. 118 up-regulated and 98 down-regulated miRNA were identified from GSE59247 dataset, and 226 up-regulated and 107 down-regulated miRNA were identified from TCGA-BRCA dataset.

Dataset Upregulation Downregulation
GSE59247 118 98
TCGA-BRCA 226 107
Table 1 Number of miRNA with significantly changed expression in breast cancer.

Venn diagram was plotted to show the total numbers of miRNA with increased expression in breast cancer from GSE59247 dataset and TCGA-BRCA dataset, and the intersection of the Venn diagram shows that 9 miRNA were identified from both datasets.

Figure 2 Venn diagram showing the number of up-regulated miRNA from GSE59247 and TCGA-BRCA datasets.

Volcano plots were generated to show the distribution of the miRNA on fold change and p-value from both GSE59247 dataset and TCGA-BRCA dataset.

Figure 3 Volcano plot of miRNA from GSE59247 dataset.
Figure 4 Volcano plot of miRNA from TCGA-BRCA dataset.

ROC Curve

To further refine the selection of target miRNAs for BreFast detection, we plotted ROC curves for the 9 miRNAs identified from differential expression analysis. Since the sample size in the GSE59247 dataset was relatively small, we used the TCGA-BRCA dataset to generate the ROC curves, as shown in the figure below.

Figure 5 ROC curves generated from TCGA-BRCA data.

The area under the curve is an important index of the ROC curve. The larger the area under the curve, the higher the sensitivity and specificity of the corresponding miRNA. AUC>0.9 was used as a criterion for selection, and miR-21-5p, miR-183-5p and miR-96-5p were selected for subsequent analysis.

Feature importance analysis

Feature importance analysis is a machine learning method to analyze how important of a feature to a certain characteristics (label). We treated each miRNA as a feature, and labeled samples as positive or negative based on whether they were from cancer patients or normal persons, thus transforming the problem of selecting optimal cancer-associated miRNA into a machine learning feature importance issue. Datasets were divided into training data and testing data in an 8:2 ratio. We used grid search and five-fold cross-validation to find the optimal parameters (achieving an accuracy of 0.98924 in five-fold cross-validation). The binary classification task was performed using a random forest model, and the accuracy reached 1 on the testing dataset. Feature importance was calculated based on the Gini index, and the top 10 ranked miRNAs were shown below.

Figure 6 The feature importance of the top ten miRNAs analyzed by random forest, where the upregulated and downregulated miRNAs have been labeled based on the results of differential expression analysis of TCGA data.

While tree models, such as random forest, can reflect the classification value of a feature (each miRNA) concerning sample (each patient) classification, they usually do not consider the interactions between different features. Considering the possibility of strong interactions among different miRNAs[1], our team has employed SHAP (SHapley Additive explanations) analysis, a methodology based on cooperative game theory, to conduct further analysis on the feature importance derived from the Random Forest model. The top 10 miRNA with high SHAP values were shown below, and miR-21-5p ranked first.

Figure 7 SHAP value analysis of the feature importance derived from Random Forest, where the upregulated and downregulated miRNAs have been labeled based on the results of differential expression analysis of TCGA data.

In summary, our bioinformatic analysis demonstrated that miR-21-5p is the optimal breast cancer-associated miRNA for detection.

Survival Analysis

Differential expression analysis and feature importance analysis have been performed to identify miR-21-5p as an optimal target for breast cancer screening. Next, we would like to know whether miR-21-5p could truly affect the progression of breast cancer. Therefore, we did survival analysis of miR-21-5p with Kaplan-Meyer curves. The data of Metabric and TCGA were used to draw the survival curve of miR-21-5p.

Figure 8 The survival curves for miR-21-5p, where Figure (a) represents data from the Metabric database, and Figure (b) represents data from the TCGA dataset.

The results of survival analysis demonstrated that the survival time of breast cancer patients with high expression of miR-21-5p was shorter than that of patients with low expression, and there was a statistically significant correlation between miR-21-5p and the survival time of breast cancer patients.

Literature Research

In addition to bioinformatics analysis, we also read literatures to confirm that miR-21-5p is the best miRNA candidate to be used as the optimal breast cancer-associated biomarkers for screening by Brefast.

A comprehensive review on International Journal of Molecular Sciences summarized 75 independent clinical studies. 20 miRNAs were suggested to be breast cancer associated biomarkers, which were supported by at least two independent studies in the same biological specimen (serum, plasma, or whole blood). 8 out 20 of them demonstrated similar tendency of change in expression: miR-21 (12 up versus 1 down), miR-155 (14 up versus 1 down), miR-10b (5 up, only serum), miR-373 (3 up, only serum), miR-652 (3 down, only serum), miR-425 (2 down, only serum), miR-29a (2 up, only serum), and miR-148b (2 down, only serum). Others miRNA, such as miR145 displayed different tendency of change in expression[2]. As a result, miR-21, miR-155, miR-10b were supported by multiple studies as highly associated with breast cancer[3-4]. It is worthy to note that the predominant form of miR-21 is miR-21-5p.

Figure 9 Pyramidal graph of the direction of miRNA expression (microRNA concentration in breast cancer cases versus controls) by type of specimens (only microRNAs that were analysed in two or more independent studies). (A) whole blood; (B) plasma; (C) all specimens; (D) serum.

In summary, both our own bioinformatic analysis and studies from multiple research groups demonstrated that miR-21-5p is the best miRNA biomarker for detection of breast cancer.

Experimental Investigation

Studies have shown that miR-21-5p can promote peritoneal metastasis in gastric cancer[5]. Therefore, we not only verified the expression of miR-21-5p in breast cancer cells, but also investigated the effects of miR-21-5p on the migration and invasion of breast cancer cells.

miR-21-5p is highly expressed in breast cancer cells

To determine the expression of miR-21-5p in breast cancer cells, we did qRT-PCR to determine the relative expression of miR-21-5p in the breast epithelial cell line MCF10A and the breast cancer cell line MCF7, which showed that the expression of miR-21-5p in breast cancer cells was higher than that in normal breast cells. (Figure 10).

Figure 10 Expression of miR-21-5p in MCF10A and MCF7 cells (A) qRT-PCR primer sequence for miR-21-5p ; (B) Raw data of qRT-PCR ; (C) miR-21-5p was highly expressed in breast cancer cells (p<0.001).

miR-21-5p enhances the migration and invasion abilities of breast cancer cells

The effect of miR-21-5p on the invasion capability of breast cancer cells was investigated by Transwell assay (Figure 11). miR-21-5p was overexpressed in breast cancer cell line MDA-MB-231 by transfection. The invasion ability of MDA-MB-231 transfected with miR-21-5p were significantly higher than that of cells transfected with control RNA, demonstrating that miR-21-5p increased the invasiveness of MDA-MB-231 cells (Figure 12).

Figure 11 Schematic diagram of Transwell invasion assay.
Figure 12 (A) Penetration of cells in the compartment; (B) Cell count results (n=5, p <0.001) .

To verify the effects of miR-21-5p on the migration of breast cancer cells, wound healing experiment was conducted. MDA-MB-231 cells were transfected with miR-21-5p or control RNA, and the migration of cells were recorded at 0h, 24h and 48h. The wound area of the MDA-MB-231 cells tranfected with miR-21-5p group was significantly smaller than that of MDA-MB-231 cells tranfected with control RNA, indicating that miR-21-5p enhanced the migration ability of MDA-MB-231 cells (Figure 13).

Figure 13 Wound healing assays of miR-21-5p transfected cells (A) Wound healing experimental images taken at 0h, 24h and 48h; (B) Relative migration level (p<0.001).

In summary, the expression level of miR-21-5p in breast cancer cell MCF7 is much higher than that in breast epithelial cell MCF10A, and increased expression of miR-21-5p enhanced the migration and invasion capability of breast cancer cells. Together, these results demonstrated that increased expression of miR-21-5p had significant effects on breast cancer cells.

LwaCas13a

Construction of pSB1C3-His-SUMO-LwaCas13a plasmid

Since we decided to use CRISPR/Cas13a system to detect miR-21-5p, we constructed a pSB1C3-His-SUMO-LwaCas13a plasmid for expression of LwaCas13a. The schematic presentation of pSB1C3-His-SUMO-LwaCas13a plasmid was shown below (Figure 14C). LacI operon was used for the induced expression of LwaCas13a[6], and a SUMO tag was added to the N-terminal of LwaCas13a protein to increase the solubility[7]. To purify the LwaCas13a protein, a 6×His tag was also added to the N-terminal of LwaCas13a protein[8].

The pSB1C3-His-SUMO-LwaCas13a plasmid was constructed using PCR amplification and Golden gate assembly technology which is an excellent methods of DNA recombination capture the principal advantages of the BioBrick standard[9]. Fragment containing SUMO-LwaCas13a was amplified from plasmid pC013-TwinStrep-SUMO-huLwCas13a by primers Cas13a F and Cas13a R. Fragment containing LacI operon was amplified from the plasmid pC013-TwinStrep-SUMO-huLwCas13a by primers LacI F and LacI R. Fragment containing pSB1C3 backbone sequence was amplified from the plasmid Pveg by primers Backbone F and Backbone R (Figure 14A,B).

Figure 14 Cloning strategy for pSB1C3-His-SUMO-LwaCas13a (A) Illustration of Pveg plasmid. (B) Illustration of pC013-Twinstrep-SUMO-huLwCas13a. (C) Illustration of pSB1C3-His-SUMO-LwaCas13a plasmid.

The PCR products were analyzed by electrophoresis with 1% agarose gel, which showed that the pSB1C3 backbone fragment (2086 bp), LacI operon fragment (1525 bp) and SUMO-LwaCas13a fragment (4227 bp) were amplified successfully (Figure 15).

Figure 15 Gel electrophoresis of the PCR product for Golden Gate assembly of pSB1C3-His-SUMO-LwaCas13a plasmid. Lane1: DNA marker. Lane2-Lane3: LwaCas13a. Lane4-Lane5: LacI operating system. Lane6-Lane7: pSB1C3 Backbone.

These fragments were assembled to generate pSB1C3-His-SUMO-LwaCas13a by Golden Gate assembly, and the assembled products were transformed into Trans1-T1 competent bacterium (Figure 16).

Figure 16 Colonies that grow after transformation.

To select colonies with successful assembly, colony PCR was performed. PCR Product 1 was amplified by Primer VR and Primer CHECK F that covered the junction between fragment SUMO-LwaCas13a and fragment LacI operon and the junction between fragment LacI operon and fragment pSB1C3 backbone (3000 bp). PCR Product 2 amplified by Primer CHECK R and Primer VF2 covered the junction between fragment pSB1C3 backbone and fragment SUMO-LwaCas13a (3050 bp). The two products were analyzed by agarose gel electrophoresis (Figure 17). Gel electrophoresis results demonstrated that the Golden Gate assembly was successful.

Figure 17 Gel electrophoresis of the colony PCR product. (A) Illustration of PCR Product 1. (B) Illustration of PCR Product 2. (C) Lane1: Marker. Lane2-Lane4: The product amplified by Primer VR and Primer CHECK F. Lane5-Lane7: The product amplified by Primer CHECK R and Primer VF2.

For further verification, we carried out the endonuclease digestion. The restriction endonuclease EcoRI was used for single cut, and the restriction endonuclease EcoRI and Spel were used for double cut, followed by 1% agarose gel electrophoresis. Two bands (5712bp and 2068bp) were produced by digestion with both EcoRI and SpeI, while one band (7780bp) was produced by digestion with EcoRI only. The electrophoresis results confirmed successful assembly of pSB1C3-His-SUMO-LwaCas13a (Figure 18).

Figure 18 Plasmid pSB1C3-His-SUMO-LwaCas13a was verified by endonuclease digestion (A) Digestion using EcoRI and SpeI results in two bands. One is 5712 bp, and the other is 2068 bp.; (B) Digestion using EcoRI enzymes results in a band with a length of 7780bp. (C) Lane1: Marker. Lane2: Plasmid was digested with EcoRI and SpeI. Lane3: Plasmid was digested with EcoRI.

Expression and purification of LwaCas13a protein

5 mL LB medium and 1 mL of bacteria-glycerol mixture were added into 15 mL tubes, and cultured on a shaker for 4 h until the bacteria culture were cloudy. They were then transferred into 1L flasks, and supplemented with 250mL of medium and 100 uL of CmR solution (50mg/mL), and incubated with shaking at 220rpm for 8 h. 1 mL of bacterial solution was collected for later use. Subsequently, 200 μL of 0.5M IPTG was added to each vial to induce protein expression and incubated overnight at 28°C. The cell lysate from bacteria before and after IPTG induction were analyzed by SDS-PAGE gel electrophoresis, and the results demonstrated that the expression of LwaCas13a was induced successfully (Figure 19).

Figure 19 Results of SDS-PAGE gel analysis of cell lysate from bacteria before and after IPTG induction. Lane1: Protein marker. Lane2-Lane6: Uninduced cell lysate from bacteria. Lane7-Lane11: IPTG-induced cell lysate from bacteria.

Bacteria were lysed by ultrasound, and centrifuged to separate supernatant and pellet. The soluble proteins were present in the supernatant, and the insoluble proteins were in the pellet. Both supernatant and pellet were analyzed by SDS-PAGE (Figure 20). SDS-PAGE gel analysis showed that the amount of LwaCas13a protein in the pellet was very little, and most of LwaCas13a protein was in the supernatant, indicating that SUMO tagged LwaCas13a was soluble.

Figure 20 SDS-PAGE detection of lysed supernatant of cells with lysis pellet of cells. Lane1: Protein marker. Lane2-Lane6: Lysed supernatant of cells. Lane7-Lane11: Lysis pellet of cells.

Western blot examined uninduced bacteria lysed by RIPA, induced bacteria lysed by RIPA, and induced bacteria lysed by ultrasonic, and showed that SUMO-LwaCas13a protein was expressed successfully (Figure 21).

Figure 21 Western Blot with LwaCas13a antibody. Lane1: Protein marker. Lane2-Lane3: Uninduced bacteria for RIPA-lysed. Lane4-Lane5: Induced bacteria for RIPA-lysis. Lane6-Lane7: Induced bacteria for ultrasonic lysis.

The final step of LwaCas13a production was to use His-Tag Protein Purification Kit with IDA-Ni Magnetic Agarose Beads for purification[10] and SUMO protease ULP1 to digest SUMO-LwaCas13a. We used SDS-PAGE gel electrophoresis to analyze the purification results (Figure 22).

Figure 22 Results of purification of LwaCas13a protein. Lane1:Protein marker. Lane2-Lane3: Uninduced bacteria for RIPA-lysed. Lane4-Lane5: Induced bacteria for RIPA-lysis. Lane6-Lane7: Induced bacteria for ultrasonic lysis. Lane8-Lane9: Purified LwaCas13a protein.

crRNA

Construction of pSB1C3-crRNA plasmid

crRNA consists of two parts, a repeat part that contains a loop region and a detection part that contains a linear guide sequence to recognize target RNA[11]. Studies have shown that the length of crRNA guide sequence affects its activity. When the length of crRNA guide sequence is less than 20nt, its activity is significantly decreased[12]. We designed four crRNA guide sequences that were complementary to miR-21-5p, the length of which ranges from 20nt to 22nt. The secondary structures of these four crRNA were as below (Figure 23).

Figure 23 The secondary structure of crRNA1 (A), crRNA2 (B), crRNA3 (C), crRNA full length (D).

crRNA sequences were cloned into Pveg plasmid with PCR. Most loop region of crRNA was synthesized as an overhang part of primer loop R, which was introduced into the fragment 1 by PCR with primer loop R and primer loop F. The rest of loop region and the guide sequence of crRNA was synthesized as an overhang part of primer spacer F, which was introduced into the fragment 2 by PCR with primer spacer F and primer spacer R. Fragment 1 and 2 were assembled into a plasmid with full length crRNA by Golden Gate assembly. (Figure 24).

Figure 24 cloning of Pveg plasmid with full-length crRNA (A) Schematic presentation of cloning strategy of Pveg plasmid with full-length crRNA sequences. (B) Primer sequences for cloning of crRNA into Pveg.

PCR was carried out, followed by gel electrophoresis, which demonstrated that fragments containing loop region and guide sequences were amplified successfully (Figure 25).

Figure 25 Gel electrophoresis of PCR products for Pveg plasmid with full-length crRNA. Lane1: DNA marker. Lane2: Guide sequence of crRNA1. Lane3: Guide sequence of crRNA2. Lane4: Guide sequence of crRNA3. Lane5: Guide sequence of crRNA full length. Lane6: Loop region.

The assembled product was transformed into Trans1-T1 competent cells, and then colony PCR was performed to select the assembled product. PCR product amplified by primer VR and VF2 covered the junction between the fragment 1 and the fragment 2 (403bp/401bp). Agarose gel electrophoresis showed that the assembly of the two fragments was successful (Figure 26).

Figure 26 PCR verified that the plasmid has been successfully assembled. (A)Schematic representation of PCR product.(B) Colony gel electrophoresis after PCR. Lane1: DNA Marker. Lane2-Lane4: crRNA full length. Lane5-Lane7: crRNA1. Lane8-Lane10: crRNA2. Lane11-Lane13: crRNA3.

In vitro transcription and purification of crRNA

Prior to in vitro transcription of crRNA, Pveg plasmids with full crRNA sequences were linearized by SapI digestion. The linearized plasmids were used for in vitro transcription using the T7 High Yield RNA Synthesis Kit from YEASEN Biotechnology Co., Ltd. The reaction system was shown in Table 2, and the mixture was incubate at 37 °C for 5h to allow the transcription reaction to proceed sufficiently.

Reagent Quantily μL(for 20μL mix)
T7 RNA Polymerase Mix 2μL
NTP Mix (100mM) 8μL
T7 Reaction Buffer (10×) 2μL
RNase Inhibitor 0.5μL
Template DNA 1μg
RNase-free water to 20μL
Total 20μL
Table 2 In vitro transcription reaction system.

Transcription products were recovered using the RNA Purification Kit from Tiangen. RK solution and absolute ethanol were added to the product, and mixed well. The mixture was added to the adsorption column and centrifuged. The column was washed with deproteinization solution and rinse solution, and elution buffer was added to obtain purified crRNA. Agarose gel electrophoresis showed that all four crRNAs were successfully transcripted and purified (Figure 27).

Figure 27 Gel electrophoresis plot of in vitro transcription of crRNA. Lane1: crRNA full length. Lane2: crRNA1.Lane3: crRNA2. Lane4: crRNA3.

In summary, we have made two essential components of CRISPR/Cas13a system: crRNA and LwaCas13a protein.

Cleavage activity

Since fluorescence reaction has the characteristics of high sensitivity and quantification[13], the cleavage activity of our CRISPR/Cas13a system was first analyzed by fluorescence detection. We used a reporter RNA designed based on the principle of fluorescence resonance energy transfer (FRET)[14], with a FAM group at one end and a black hole quencher group (BHQ1) at the other end, connected by 6 U in between. Activated CRISPR/Cas13a system could cleave the reporter at 6 U, and release FAM group from the quencher group. As a result, fluorescence from the released FAM group could be detected. If the crRNA that we designed can form a functional complex with LwaCas13a, the cleavage activity of this complex could be activated by miR-21-5p, which could cleave the reporter RNA to release FAM group for fluorescence detection.

Four crRNA named crRNA1, crRNA2, crRNA3 and crRNA full length, were used in the experiment. The results showed that the CRISPR reaction system assembled by crRNA full length and Cas13a protein had the strongest cleavage activity, followed by crRNA3, while crRNA2 and crRNA3 had the weakest activity (Figure 28).

Figure 28 Fluorescence intensity measured at different time with different crRNAs.

Next, for the fluorescence reaction, the concentrations of crRNA, LwaCas13a, and reporter RNA were fixed at 20 nM, 100 nM and 100 nM respectively, and the concentration of miR-21-5p was gradually increased from 0 nM to 100 nM. The experimental results showed that the fluorescence intensity increased as the concentration of the miRNA increased (Figure 29).

Figure 29 Fluorescence intensity measured at different times with different concentrations of miRNA.

In addition, we also set up a series of reporter concentration at 0 nM, 100 nM, 125 nM, 250 nM and 500 nM for experiments. After we plotted the double inverse of fluorescence intensity and reporter RNA concentration, the experimental results were found to be consistent with Michaelis-Menten equation[15], which was in line with our expectations. (Figure 30).

Figure 30 (A) Fluorescence intensity measured at different times with different concentrations of reporter RNA. (B) The fitting curve of Lineweaver-Burk.

Lateral flow strip

After completing a series of fluorescence experiments that validated the cleavage activity of our CRISPR/Cas13a system, we began to simplify the method that was used to detect the cleavage activity of the CRISPR/Cas13a system with lateral flow strips.

To facilitate the lateral flow strip to display the results, the BHQ1 modification of our RNA reporter 3' was changed to biotin, while the sequence remained unchanged, still 6 U. In the experiment of lateral flow strip test, we adopted the detection method of "line elimination". On the probe pad of the lateral flow strip, there is streptavidin-labeled colloidal gold, which binds to the biotin at the RNA reporter. The position of the Test line (T line) at the lateral flow strip is planted with FAM antibodies, which binds to FAM group of the RNA reporter. In the case that the RNA reporter is uncleaved, the RNA reporter forms a complex with the streptavidin-labeled colloidal gold through the interaction between biotin and streptavidin. As this complex flows to the T line, it is captured by the FAM antibody that binds to the FAM group of the RNA reporter. As a result, the colloidal gold accumulates at the T line, which can be observed by naked eyes. In addition, the gold-labeled mouse antibodies are included in the lateral flow strip detection system as controls. They are captured by the sheep-anti mouse antibodies at the C line position to show the samples are added successfully on the lateral flow strip.

In the case that miR-21-5p is not expressed, the CRISPR/Cas13a system is not activated, and the RNA reporter is not cleaved. The uncleaved RNA reporter with streptavidin-labeled colloidal gold is captured by the FAM antibody at the T-line position, and the gold labeled mouse antibodies is captured by the sheep-anti mouse antibodies at the C line position. As a result, the lateral flow strip shows both C line and T line, indicating negative result (Figure 31).

In the case that miR-21-5p is expressed low, the CRISPR/Cas13a system is not fully activated, the reporter is not completely cleaved. The uncleaved RNA reporter with streptavidin-labeled colloidal gold can still be captured by the FAM antibody at the T-line position, while the cleaved RNA reporter can not link streptavidin-labeled colloidal gold with FAM antibody anymore. As a result, the lateral flow strip shows C line and a weak T line, indicating negative result.

Figure 31 Principle of our lateral flow strips.

Next, the lateral flow strip was tested. The CRISPR/Cas13a system was composed of working solution and sample solution, and they were mixed for reaction. The composition of the working solution included 10×Cas13a Reaction Buffer (the recipe was shown in Table 3), RNase Inhibitor, LwaCas13a protein, crRNA-Full length and RNase-free water, and it has a total volume of 10μL (Table 4). The composition of the sample solution includes miR-21-5p, reporter and so on, its volume was also 10μL (Table 5). The concentration of miRNA in the reaction system was changed to test the effects of strips.

Component Concentration
Tris-HCl 10mM
KCl 50nM
MgCl2 1.5mM
pH 8.3
Table 3 Components of Cas13a Reaction Buffer (10×).
Reagent Quantily μL (for 10μL mix)
Cas13a Reaction Buffer (10×) 2μL
RNase Inhibitor 1μL
LwaCas13a (2μM) 1μL
crRNA full length (0.4μM) 1μL
RNase-free water 5μL
Total 10μL
Table 4 Components of the working solution.
Reagent Quantily μL (for 10μL mix)
Cas13a Reaction Buffer (10×) 2μL
RNase Inhibitor 1μL
miR-21-5p (1μM) xμL
Reporter (2μM) 2.4μL
RNase-free water to 10μL
Total 10μL
Table 5 Components of the sample solution.

After mixing the working solution and the sample solution, the reaction solution was obtained (Figure 32), and the reaction droplets were added to the sample pad of the test strip, the result was recorded in 3 minutes.

Figure 32 The reaction solution with different concentration of miRNA.

The results of the lateral flow strip detection were as in Figure 33. The C lines of the five test strips were clearly visible, indicating that these test strips were good to use. In the case that the miRNA concentration in the reaction mix was 0 nM, T line was obvious. While the miRNA concentration increased to 5 nM, T line was decreased. A further increase of miRNA concentration to 25 nM made the T line almost disappeared. In the case that the miRNA concentration increased to 50 and 100 nM, T line completely disappeared. These results demonstrated that the lateral flow worked as we expected, which could distinguish low and high concentration of miR-21-5p easily (Figure 33).

Figure 33 High and low concentrations of miRNA were distinguished successfully by lateral flow strip.

Subsequently, the outer shell of the lateral flow strip was manufactured to make it easy to carry and work with (Figure 34-35).

Figure 34 Diagram of the lateral flow strip and its outer shell.
Figure 35 The outer shell of the lateral flow strip facilitates sample loading and result interpretation.

Future plan

In this year's project, we engineered a CRISPR/Cas13a system to detect miR-21-5p for breast cancer screening. In the future, we set to make a breast cancer screening kit that can simultaneously detect multiple breast cancer-associated miRNAs, such as miR-21-5p, miR-3923, miR-885-5p and miR-142-5p, so as to achieve higher sensitivity and accuracy of breast cancer detection. In addition, we would like to apply our CRISPR/Cas13a system to screening of other types of cancers.

References

[1] https://www.datacamp.com/tutorial/introduction-to-shap-values-machine-learning-interpretability.

[2] Padroni, L., et al. (2023). "An Epidemiological Systematic Review with Meta-Analysis on Biomarker Role of Circulating MicroRNAs in Breast Cancer Incidence." Int J Mol Sci 24(4).

[3] Weng, S., et al. (2022). "Highly sensitive and reliable detection of microRNA for clinically disease surveillance using SERS biosensor integrated with catalytic hairpin assembly amplification technology." Biosens Bioelectron 208: 114236.

[4] Fan, T., et al. (2018). "Branched rolling circle amplification method for measuring serum circulating microRNA levels for early breast cancer detection." Cancer Sci 109(9): 2897-2906.

[5] Li, Q., et al. (2018). "Exosomal miR-21-5p derived from gastric cancer promotes peritoneal metastasis via mesothelial-to-mesenchymal transition." Cell Death Dis 9(9): 854.

[6] Kercher, M. A., et al. (1997). "Lac repressor-operator complex." Curr Opin Struct Biol 7(1): 76-85.

[7] Kuo, D., et al. (2014). "SUMO as a solubility tag and in vivo cleavage of SUMO fusion proteins with Ulp1." Methods Mol Biol 1177: 71-80.

[8] Tropea, J. E., et al. (2009). "Expression and purification of soluble His(6)-tagged TEV protease." Methods Mol Biol 498: 297-307.

[9] Matsumura, I. (2022). "Golden Gate assembly of BioBrick-compliant parts using Type II restriction endonucleases." Biotechniques 72(5): 185-193.

[10] Wang, J., et al. (2021). "Phase separation of OCT4 controls TAD reorganization to promote cell fate transitions." Cell Stem Cell 28(10): 1868-1883.e1811.

[11] Kellner, M. J., et al. (2019). "SHERLOCK: nucleic acid detection with CRISPR nucleases." Nat Protoc 14(10): 2986-3012.

[12] Abudayyeh, O. O., et al. (2017). "RNA targeting with CRISPR-Cas13." Nature 550(7675): 280-284.

[13] Shan, Y., et al. (2019). "High-Fidelity and Rapid Quantification of miRNA Combining crRNA Programmability and CRISPR/Cas13a trans-Cleavage Activity." Analytical Chemistry 91(8): 5278-5285.

[14] Jares-Erijman, E. A. and T. M. Jovin (2003). "FRET imaging." Nat Biotechnol 21(11): 1387-1395.

[15] Ramachandran, A. and J. G. Santiago (2021). "CRISPR Enzyme Kinetics for Molecular Diagnostics." Anal Chem 93(20): 7456-7464.