[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,518)

Search Parameters:
Keywords = CRISPR

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
43 pages, 1332 KiB  
Review
Bioremediation of Smog: Current Trends and Future Perspectives
by Isha, Shakir Ali, Ammara Khalid, Ifrah Amjad Naseer, Hassan Raza and Young-Cheol Chang
Processes 2024, 12(10), 2266; https://doi.org/10.3390/pr12102266 - 17 Oct 2024
Viewed by 381
Abstract
Air pollution has become one of the biggest problems throughout the world. Smog has a severe effect on the pulmonary and circulatory systems, which causes a significant number of deaths globally. Therefore, the remediation of air pollutants to maintain ecosystem processes and functions [...] Read more.
Air pollution has become one of the biggest problems throughout the world. Smog has a severe effect on the pulmonary and circulatory systems, which causes a significant number of deaths globally. Therefore, the remediation of air pollutants to maintain ecosystem processes and functions and to improve human health is a crucial problem confronting mankind today. This review aims to discuss the health effects of smog on humans. This review will also focus on the bioremediation of air pollution (smog) using bacteria, fungi, phytoremediation, nanotechnology, and phylloremediation (using plants and microbes). Phylloremediation is the most effective technology for removing air pollution naturally. The future perspective presents a great need to produce an ecosystem where microbes, plants, and nanoparticles synergistically control smog. In addition, further advancements would be needed to modify the genetic makeup of microbes and plants. Biotechnological approaches like CRISPR-Cas9 can be applied to the editing and cutting of specific genes responsible for the bioremediation of VOCs, NOx, SOx, and harmful hydrocarbons. The extracted genes can then be expressed in biologically modified microorganisms and plants for the enhanced bioremediation of smog. Full article
(This article belongs to the Special Issue Advanced Biodegradation Technologies for Environmental Pollutants)
Show Figures

Figure 1

Figure 1
<p>The primary systems of the human body are mainly affected by smog, i.e., the respiratory system, the circulatory system, and the nervous system, leading to asthma and neuronal cell death.</p>
Full article ">Figure 2
<p>Smog pollutants, health effects, and prevalence in Punjab (WHO). Prevalence indicates deaths; PM (particulate matter); POCs (persistent organic compounds); and NOx (nitrogen oxides); VOCs (volatile organic compounds).</p>
Full article ">Figure 3
<p>Types of bioremediation and the common methods followed by in situ and ex situ bioremediation.</p>
Full article ">
12 pages, 2742 KiB  
Article
Effects of OsLPR2 Gene Knockout on Rice Growth, Development, and Salt Stress Tolerance
by Ying Gu, Chengfeng Fu, Miao Zhang, Changqiang Jin, Yuqi Li, Xingyu Chen, Ruining Li, Tingting Feng, Xianzhong Huang and Hao Ai
Agriculture 2024, 14(10), 1827; https://doi.org/10.3390/agriculture14101827 - 17 Oct 2024
Viewed by 321
Abstract
Rice (Oryza sativa L.), a globally staple food crop, frequently encounters growth, developmental, and yield limitations due to phosphate deficiency. LOW PHOSPHATE ROOT1/2 (LPR1/2) are essential genes in plants that regulate primary root growth and respond [...] Read more.
Rice (Oryza sativa L.), a globally staple food crop, frequently encounters growth, developmental, and yield limitations due to phosphate deficiency. LOW PHOSPHATE ROOT1/2 (LPR1/2) are essential genes in plants that regulate primary root growth and respond to local phosphate deficiency signals under low phosphate stress. In rice, five LPR genes, designated OsLPR1OsLPR5 based on their sequence identity with AtLPR1, have been identified. OsLPR3 and OsLPR5 are specifically expressed in roots and induced by phosphate deficiency, contributing to rice growth, development, and the maintenance of phosphorus homeostasis under low phosphate stress. In contrast, OsLPR2 is uniquely expressed in shoots, suggesting it may have distinct functions compared with other family members. This study employed Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-associated protein 9 (CRISPR/Cas9) gene editing technology to generate oslpr2 mutant transgenic lines and subsequently investigated the effect of OsLPR2 gene knockout on rice growth, phosphate utilization, and salt stress tolerance in the seedling stage, as well as the effect of OsLPR2 gene knockout on rice development and agronomic traits in the maturation stage. The results indicated that the knockout of OsLPR2 did not significantly impact rice seedling growth or phosphate utilization, which contrasts significantly with its homologous genes, OsLPR3 and OsLPR5. However, the mutation influenced various agronomic traits at maturity, including plant height, tiller number, and seed setting rate. Moreover, the OsLPR2 mutation conferred enhanced salt stress tolerance in rice. These findings underscore the distinct roles of OsLPR2 compared with other homologous genes, establishing a foundation for further investigation into the function of the OsLPR family and the functional differentiation among its members. Full article
(This article belongs to the Section Crop Genetics, Genomics and Breeding)
Show Figures

Figure 1

Figure 1
<p>Transcript level of <span class="html-italic">OsLPR2</span> under different nutrient deficiencies. Wild type rice seedlings of Nipponbare were cultivated for 7 days in complete nutrient solution (CK) or in nutrient-deficiency solution, which excluded nitrogen (−N), phosphorus (−P), potassium (−K), magnesium (−Mg), or iron (−Fe). Relative expression levels of OsLPR2 in shoot (<b>A</b>) and root (<b>B</b>) were determined via qRT-PCR. Values are presented as means ± SE (<span class="html-italic">n</span> = 3). Different letters above the bars indicate significant differences in the relative expression levels of OsLPR2 (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA).</p>
Full article ">Figure 2
<p>Construction and identification of <span class="html-italic">OsLPR2</span> mutant material. (<b>A</b>) Schematic diagram of the <span class="html-italic">oslpr2</span> target sites. (<b>B</b>) Identification of positive <span class="html-italic">oslpr2</span> seedlings. (<b>C</b>) Sequencing sequences and chromatograms of homozygous <span class="html-italic">oslpr2</span> mutant lines. (<b>D</b>) Cas9 segregation identification of mutant lines.</p>
Full article ">Figure 3
<p>The effect of <span class="html-italic">OsLPR2</span> mutation on the plant height and tillers per plant at maturity. (<b>A</b>) Plant types. (<b>B</b>) Plant height. (<b>C</b>) Number of tillers per plant. Scale bar: 20 cm. Values are means ± SE (<span class="html-italic">n</span> = 15). Different letters above the bars indicate significant differences (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA).</p>
Full article ">Figure 4
<p>The effect of <span class="html-italic">OsLPR2</span> mutation on panicle type of rice. (<b>A</b>) Panicle types. (<b>B</b>) Panicle length. (<b>C</b>) Number of primary branches. (<b>D</b>) Number of secondary branches. (<b>E</b>) Number of grains per panicle. (<b>F</b>) Seed setting rate. Scale bar: 5 cm. Values are means ± SE (<span class="html-italic">n</span> = 15). Different letters above the bars indicate significant differences (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA).</p>
Full article ">Figure 5
<p>The effect of <span class="html-italic">OsLPR2</span> mutation on the lengths of shoots and roots. (<b>A</b>,<b>B</b>) Images showing the relative growth performances of WT and <span class="html-italic">oslpr2</span> mutant lines under +P and −P conditions (bar = 10 cm). (<b>C</b>,<b>E</b>) Lengths and biomass of shoots or roots under phosphate sufficiency. (<b>D</b>,<b>F</b>) Lengths and biomass of shoots and roots under phosphate deficiency. Values are presented as means ± SE (<span class="html-italic">n</span> = 6). Same letters above the bars indicate no significant differences (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA).</p>
Full article ">Figure 6
<p>The effect of <span class="html-italic">OsLPR2</span> mutation on soluble Pi concentration of rice. (<b>A</b>) <span class="html-italic">OsLPR2</span> transgenic materials and wild type plants with consistent growth under normal phosphate supply. (<b>B</b>) After 21 days of phosphate deficiency treatment, sampling of different plant parts (leaves, leaf sheaths, roots) for extractable phosphate content measurement. Values are means ± SE (<span class="html-italic">n</span> = 3). Different letters above the bars indicate significant differences (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA).</p>
Full article ">Figure 7
<p>Assessment of <span class="html-italic">oslpr2</span> mutant survival and physiological responses under saline conditions. (<b>A</b>) Phenotypes of WT and <span class="html-italic">oslpr2</span> mutants after 200 mM NaCl treatment. (<b>B</b>) Survival rate statistics. (<b>C</b>) POD activity after 150 mM NaCl treatment. (<b>D</b>) MDA content after 150 mM NaCl treatment. Values are means ± SE (<span class="html-italic">n</span> = 3). Different letters above the bars indicate significant differences (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA).</p>
Full article ">
17 pages, 4853 KiB  
Article
The Aspergillus flavus hacA Gene in the Unfolded Protein Response Pathway Is a Candidate Target for Host-Induced Gene Silencing
by Perng-Kuang Chang
J. Fungi 2024, 10(10), 719; https://doi.org/10.3390/jof10100719 - 16 Oct 2024
Viewed by 339
Abstract
Fungal HacA/Hac1 transcription factors play a crucial role in regulating the unfolded protein response (UPR). The UPR helps cells to maintain endoplasmic reticulum (ER) protein homeostasis, which is critical for growth, development, and virulence. The Aspergillus flavus hacA gene encodes a domain rich [...] Read more.
Fungal HacA/Hac1 transcription factors play a crucial role in regulating the unfolded protein response (UPR). The UPR helps cells to maintain endoplasmic reticulum (ER) protein homeostasis, which is critical for growth, development, and virulence. The Aspergillus flavus hacA gene encodes a domain rich in basic and acidic amino acids (Bsc) and a basic leucine zipper (bZip) domain, and features a non-conventional intron (Nt20). In this study, CRISPR/Cas9 was utilized to dissect the Bsc-coding, bZip-coding, and Nt20 sequences to elucidate the relationship between genotype and phenotype. In the Bsc and bZip experimental sets, all observed mutations in both coding sequences were in frame, suggesting that out-of-frame mutations are lethal. The survival rate of transformants in the Nt20 experiment set was low, at approximately 7%. Mutations in the intron primarily consisted of out-of-frame insertions and deletions. In addition to the wild-type-like conidial morphology, the mutants exhibited varied colony morphologies, including sclerotial, mixed (conidial and sclerotial), and mycelial morphologies. An ER stress test using dithiothreitol revealed that the sclerotial and mycelial mutants were much more sensitive than the conidial mutants. Additionally, the mycelial mutants were unable to produce aflatoxin but still produced aspergillic acid and kojic acid. RNAi experiments targeting the region encompassing Bsc and bZip indicated that transformant survival rates generally decreased, with a small number of transformants displaying phenotypic changes. Defects in the hacA gene at the DNA and transcript levels affected the survival, growth, and development of A. flavus. Thus, this gene may serve as a promising target for future host-induced gene-silencing strategies aimed at controlling infection and reducing aflatoxin contamination in crops. Full article
(This article belongs to the Special Issue Mycotoxin Contamination and Control in Food)
Show Figures

Figure 1

Figure 1
<p>The <span class="html-italic">A. flavus hacA</span> gene. (<b>A</b>) Schematic representation of the functional domain-coding regions and introns. Gray and black areas indicate the single conventional intron (I54) and the non-conventional intron (I20 = Nt20). BA indicates the coding region rich in basic and acidic amino acids, which is interrupted by the conventional intron. bZip denotes the basic leucine zipper coding region. (<b>B</b>) Comparison of the 20-nucleotide non-conventional introns of <span class="html-italic">A. flavus</span>, <span class="html-italic">A. parasiticus</span>, <span class="html-italic">A. oryzae</span>, <span class="html-italic">A. fumigatus</span>, <span class="html-italic">A. nidulans</span>, <span class="html-italic">A. niger</span>, <span class="html-italic">T. reesei</span>, and <span class="html-italic">V. dahlia</span>. The hexanucleotide repeat, which is the <span class="html-italic">Pst</span>I restriction site sequence, is underlined. Single nucleotide polymorphisms are highlighted. (<b>C</b>) The 3D models of HacA and HacA<sup>∆</sup>. The amino acid sequence from residues 83 to 146 is the bZip domain, which is the long α-helical structure in both forms. AlphaFold produced a colored confidence score for each residue in the α-helical structure. The score range of each color is blue &gt; 90, light blue &gt; 70 but &lt;90, yellow &gt; 50 but &lt;70, orange &lt; 50. Refer to <a href="#app1-jof-10-00719" class="html-app">Figure S3A</a> for both amino acid sequences.</p>
Full article ">Figure 2
<p>Phenotypic changes resulting from sequence mutations in the <span class="html-italic">hacA</span> functional domain-coding regions and the nonconventional intron. (<b>A</b>) The basic/acidic domain: K (lysine) and R (arginine) are basic amino acids, while E (glutamic acid) and D (aspartic acid) are acidic amino acids. The three target regions are underlined. (<b>B</b>) The bZip domain: The domain forms an α-helix (see <a href="#jof-10-00719-f002" class="html-fig">Figure 2</a>) with leucine (L) residues highlighted in blue. The three target regions are underlined. The wild-type (WT) CA14 strain is shown alongside the CS mutant for visual comparison and consistency in presentation. (<b>C</b>) The non-conventional intron: Both amino acid sequences of the wild-type CA14 strain before and after the removal of the 20-nucleotide intron are shown. Only the predicted amino acid sequences before the removal of the respective introns from the mutants with single-nucleotide insertion or deletion are shown; the removal of these introns yielded amino acid sequences identical to that of the truncated HacA<sup>∆</sup>. Single amino acid changes are highlighted in yellow. Missing amino acids are indicated by dashes, and inserted amino acids are highlighted in grey. A star indicates the end of an amino acid sequence. In the three panels, open triangles and solid inverted triangles followed by numbers indicate deleted and inserted nucleotides, respectively. The double-headed arrow indicates nucleotide substitution. The numbers after the equals signs are the number of sequenced transformants with the same mutation. The following designations are used for colony morphology: C, conidial; S, sclerotial; CS, a roughly equal mix of conidial and sclerotial, Cs, more conidial than sclerotia, Sc, more sclerotial than conidial, and M, mycelial. Mycelial mats barely contained conidiophores. The powdery-looking sectors in panels (<b>A</b>–<b>C</b>) when examined under a dissecting microscope, were found to contain aggregations of hyphae, which appeared to be white sclerotia initials.</p>
Full article ">Figure 2 Cont.
<p>Phenotypic changes resulting from sequence mutations in the <span class="html-italic">hacA</span> functional domain-coding regions and the nonconventional intron. (<b>A</b>) The basic/acidic domain: K (lysine) and R (arginine) are basic amino acids, while E (glutamic acid) and D (aspartic acid) are acidic amino acids. The three target regions are underlined. (<b>B</b>) The bZip domain: The domain forms an α-helix (see <a href="#jof-10-00719-f002" class="html-fig">Figure 2</a>) with leucine (L) residues highlighted in blue. The three target regions are underlined. The wild-type (WT) CA14 strain is shown alongside the CS mutant for visual comparison and consistency in presentation. (<b>C</b>) The non-conventional intron: Both amino acid sequences of the wild-type CA14 strain before and after the removal of the 20-nucleotide intron are shown. Only the predicted amino acid sequences before the removal of the respective introns from the mutants with single-nucleotide insertion or deletion are shown; the removal of these introns yielded amino acid sequences identical to that of the truncated HacA<sup>∆</sup>. Single amino acid changes are highlighted in yellow. Missing amino acids are indicated by dashes, and inserted amino acids are highlighted in grey. A star indicates the end of an amino acid sequence. In the three panels, open triangles and solid inverted triangles followed by numbers indicate deleted and inserted nucleotides, respectively. The double-headed arrow indicates nucleotide substitution. The numbers after the equals signs are the number of sequenced transformants with the same mutation. The following designations are used for colony morphology: C, conidial; S, sclerotial; CS, a roughly equal mix of conidial and sclerotial, Cs, more conidial than sclerotia, Sc, more sclerotial than conidial, and M, mycelial. Mycelial mats barely contained conidiophores. The powdery-looking sectors in panels (<b>A</b>–<b>C</b>) when examined under a dissecting microscope, were found to contain aggregations of hyphae, which appeared to be white sclerotia initials.</p>
Full article ">Figure 3
<p>DTT induced ER stress on the growth and development of the wild-type CA14 strain and the <span class="html-italic">hacA</span> mutants. Open triangles and solid inverted triangles followed by numbers indicate deleted and inserted nucleotides, respectively. Bsc, bZip, and Nt20 have been added to strain designations in reference to the mutated regions. For details regarding the morphologies (C, CS, S, Cs, Sc, and M) of the Bsc, bZip, Nt20 mutants, refer to the legend for <a href="#jof-10-00719-f003" class="html-fig">Figure 3</a>. Conidia from wild-type CA14 and various types of the <span class="html-italic">hacA</span> mutants were inoculated onto PDA and CZ plates, both with and without the addition of 10 mM DTT. Plates were incubated at 30 °C for four days in the dark.</p>
Full article ">Figure 4
<p>Production of aspergillic acid, anthraquinones, and kojic acid by the <span class="html-italic">hacA</span> mutants. Colony morphologies and pigmentation were examined on ADM, CAM, and KAM plates. F, top view; R, reverse side; W, white light; UV, longwave ultraviolet light. Pigmentation in ADM was restricted to the edge of the colony, while pigmentation on KAM (complex formed by diffusible kojic acid with ferric ion) was distributed throughout the plate. WT, wild-type CA14; KO, respective gene knockout mutants. Open triangles and solid inverted triangles followed by numbers indicate deleted and inserted nucleotides, respectively. Bsc, bZip, and Nt20 added to strain designations for reference to the mutated regions. Refer to the legend for <a href="#jof-10-00719-f003" class="html-fig">Figure 3</a> for strain details and designations of morphologies.</p>
Full article ">Figure 5
<p>Semi-quantitative TLC analysis of aflatoxin production by selected <span class="html-italic">hacA</span> mutants with varying colony morphologies. (<b>A</b>) The designations Bsc, bZip, and Nt20 refer to mutants with defects in their HacA functional domains and the non-conventional intron. WT denotes the wild-type CA14 strain. Open triangles and solid inverted triangles followed by numbers indicate deleted and inserted nucleotides, respectively. Refer to the legend for <a href="#jof-10-00719-f003" class="html-fig">Figure 3</a> for strain details and designations of morphologies. (<b>B</b>) Lack of aflatoxin production by the fifth transfered <span class="html-italic">hacA</span> mutant of bZip∆42/M. The mutant was serially transferred onto PDA. A small piece of mycelia-containing agar from the fourth culture (circled red) was transferred onto a PDA plate. Two agar plugs from the fifth culture were cored and analyzed using semi-quantitative TLC. Bsc∆21/M, a mycelial mutant that does not produce aflatoxin, served as a negative control.</p>
Full article ">Figure 6
<p>Colony morphology of <span class="html-italic">hacA</span>i transformants on PDA plates. Transformants exhibiting sclerotial, green and velvet-like, or retarded growth morphologies are indicated by arrows. L and R denote transformants from the L_bZip and R_bZip control sets. Cultures were incubated at 30 °C for five days in the dark.</p>
Full article ">
14 pages, 8195 KiB  
Article
The Application of Duck Embryonic Fibroblasts CCL-141 as a Cell Model for Adipogenesis
by Dan-Dan Sun, Xiao-Qin Li, Yong-Tong Liu, Meng-Qi Ge and Zhuo-Cheng Hou
Animals 2024, 14(20), 2973; https://doi.org/10.3390/ani14202973 (registering DOI) - 15 Oct 2024
Viewed by 352
Abstract
The duck embryo fibroblast cell line CCL-141, which is currently the only commercialized duck cell line, has been underexplored in adipogenesis research. (1) Background: This study establishes an experimental protocol to induce adipogenesis in CCL-141 cells, addressing the importance of understanding gene functions [...] Read more.
The duck embryo fibroblast cell line CCL-141, which is currently the only commercialized duck cell line, has been underexplored in adipogenesis research. (1) Background: This study establishes an experimental protocol to induce adipogenesis in CCL-141 cells, addressing the importance of understanding gene functions in this process. (2) Methods: Chicken serum, fatty acids, insulin, and all-trans retinoic acid were used to treat CCL-141 cells, with adipogenesis confirmed by Oil Red O staining and gene expression quantification. CRISPR/Cas9 technology was applied to knockout PPARγ, and the resulting adipogenic phenotype was assessed. (3) Results: The treatments promoted adipogenesis, and the knockout of PPARγ validated the cell line’s utility for gene function studies. (4) Conclusions: CCL-141 cells are a suitable model for investigating duck adipogenesis, contributing to the understanding of regulatory factors in this biological process. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Establishment of duck <span class="html-italic">PPARγ</span>-knockout pooled cells using CRISPR/Cas9 system. (<b>A</b>) Structure of <span class="html-italic">PPARγ</span> and location of target sequence. (<b>B</b>) Analysis of knockout efficiency using website analysis based on Sanger sequencing peak graph. (<b>C</b>) Analysis of <span class="html-italic">PPARγ</span> knockout efficiency using single cloning. Red font corresponds to target sequence. Deleted nucleotides are marked with dashes, inserted nucleotides are represented with caret “^”, mutational nucleotides are represented with lowercase letters, and protospacer adjacent motif (PAM) sequence is indicated with italics and underlined.</p>
Full article ">Figure 2
<p>Inducing effects of different culture medium components on CCL-141 cells. (<b>A</b>) Representative images of Oil Red O staining after 72 h of induction for different culture groups: (a) EMEM with 10% FBS as control; (b) EMEM with 10% chicken serum; (c) EMEM with 10% CS, 1:100 fatty acids, and 10 ug/mL insulin; and (d) EMEM with 10% CS, 1:100 fatty acids, 10 ug/mL insulin, and 40 ug/mL all-trans retinoic acid. (<b>B</b>) Comparison of lipid droplet content in different groups extracted after Oil Red O staining (different lowercase letters on columns indicate significant differences, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Differential expression of marker genes for adipogenesis in differentiating groups induced by culture medium containing different components for 48 h and 72 h (different lowercase letters indicate significant differences, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Differential expression of marker genes for adipogenesis in wild-type and knockout groups before and after 72 h of induction (different lowercase letters indicate significant differences, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Differences in protein expression level and lipid deposition in wild-type (WT) and <span class="html-italic">PPARγ</span>-KO pooled cells. (<b>A</b>) Western blotting analysis of CCL-141. Protein samples of WT and <span class="html-italic">PPARγ</span>-KO pooled cells were extracted and Western blot analysis was performed against <span class="html-italic">PPARγ</span> antibody as per procedure described in “Materials and Methods” section. (<b>B</b>) Gray value analysis of protein expression level in wild-type and <span class="html-italic">PPARγ</span>-KO pooled cells (** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>) Representative images of Oil Red O staining after 72 h of induction in wild-type and knockout groups. (<b>D</b>) Comparison of lipid droplet content in different groups extracted after Oil Red O staining (different lowercase letters indicate significant differences, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Cell proliferation curves of wild-type and knockout groups at different time points (** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
12 pages, 3468 KiB  
Article
Development of an RPA-CRISPR/Cas12a Assay for Rapid and Sensitive Diagnosis of Plant Quarantine Fungus Setophoma terrestris
by Peng Zhao, Zhipeng Feng, Lei Cai, Dorji Phurbu, Weijun Duan, Fuhong Xie, Xuelian Li and Fang Liu
J. Fungi 2024, 10(10), 716; https://doi.org/10.3390/jof10100716 - 15 Oct 2024
Viewed by 385
Abstract
Setophoma terrestris is an important phytopathogenic fungus listed by China as a harmful fungus subject to phytosanitary import control. This pathogen is a threat to a wide range of plants, particularly as the causal agent of onion pink root rot, one of the [...] Read more.
Setophoma terrestris is an important phytopathogenic fungus listed by China as a harmful fungus subject to phytosanitary import control. This pathogen is a threat to a wide range of plants, particularly as the causal agent of onion pink root rot, one of the most severe diseases of onions. In order to provide rapid identification and early warning of S. terrestris and prevent its spread, we have developed a rapid, accurate, and visually intuitive diagnostic assay for this pathogen, by utilizing recombinase polymerase amplification (RPA), coupled with CRISPR/Cas12a cleavage and fluorescence-based detection systems or paper-based lateral flow strips. The developed RPA-CRISPR/Cas12a assay exhibited remarkable specificity for the detection of S. terrestris. Moreover, this protocol can detect the pathogen at a sensitivity level of 0.01 pg/μL, which significantly outperforms the 1 pg/μL sensitivity achieved by the existing qPCR-based detection method. The entire diagnostic procedure, including DNA extraction, the RPA reaction, the Cas12a cleavage, and the result interpretation, can be accomplished in 40 min. Furthermore, the successful application of the assay in infected plant samples highlighted its potential for rapid and accurate pathogen detection in agricultural settings. In summary, this RPA-CRISPR/Cas12a diagnostic method offers a potentially valuable technological solution for quarantine and disease management. Full article
Show Figures

Figure 1

Figure 1
<p>Alignment of ITS sequences of <span class="html-italic">Setophoma</span> species. (<b>a</b>) Location of RPA primers (blue arrows) and crRNA binding sites (in yellow) recognized by PAM region (in red) in ITS region of <span class="html-italic">S. terrestris</span> were labeled. The direction of blue arrow indicates the direction of the RPA primers. (<b>b</b>) Detailed schematic of how <span class="html-italic">S. terrestris</span> crRNA and target gene fragments bind.</p>
Full article ">Figure 2
<p>(<b>a</b>–<b>d</b>) Specificity of RPA and RPA-CRISPR/Cas12a primer for <span class="html-italic">S. terrestris.</span> (<b>a</b>) RPA detection assay. (<b>b</b>) RPA-CRISPR/Cas12a-FRB. (<b>c</b>) RPA-CRISPR/Cas12a-PLFS assay. (<b>d</b>) Schematic diagram of test strip test results, T: test band, C: control band, Ne: negative. M: 100 bp DNA ladder; 1–9: <span class="html-italic">S. endophytica</span>, <span class="html-italic">S. endophytica</span>, <span class="html-italic">S. antiqua</span>, <span class="html-italic">S. longinqua</span>, <span class="html-italic">S. yunnanensis</span>, <span class="html-italic">S. caverna</span>, <span class="html-italic">S. yingyisheniae</span>, <span class="html-italic">S. terrestris</span>, <span class="html-italic">S. terrestris</span>; N: ddH<sub>2</sub>O. (<b>e</b>) Optimization of RPA reaction time. 1–5: 10 min, 20 min, 25 min, 30 min, 40 min; N: ddH<sub>2</sub>O. (<b>f</b>) Optimization of RPA reaction temperature. 1–5: 33 °C, 35 °C, 37 °C, 39 °C, 41 °C; N: ddH<sub>2</sub>O.</p>
Full article ">Figure 3
<p>(<b>a</b>–<b>d</b>) RPA-CRISPR/Cas12a-FBR assay for <span class="html-italic">S. terrestris</span>. (<b>a</b>) Optimization of Cas12a/CrRNA concentrations for visual detection and their real-time fluorescent signals. 1–5: The ratio of Cas12a/crRNA: 0 nM/0 nM, 50 nM/100 nM, 100 nM/200 nM, 150 nM/250 nM, 200 nM/300 nM; 1N–5N: ddH<sub>2</sub>O. (<b>b</b>) Optimization of reaction time for visual detection and their real-time fluorescent signals. 1–5: 5 min, 10 min, 15 min, 20 min, 30 min; 1N–5N: ddH<sub>2</sub>O. The bar chart represents data obtained from three repeated experiments, different lowercase letters denote significant differences between the groups. (<b>c</b>) Optimization of FQ-DNA reporter molecule concentrations in the reaction system and their real-time fluorescent signals. 1–5: 50 nmol/L, 100 nmol/L, 200 nmol/L, 400 nmol/L, 800 nmol/L; 1N–5N: ddH<sub>2</sub>O. (<b>d</b>) Sensitivity verification of RPA-CRISPR/Cas12a-FBR visual detection system. 1–6: 1 ng, 0.1 ng, 10 pg, 1 pg, 0.1 pg, 0.01 pg. N: ddH<sub>2</sub>O. (<b>e</b>–<b>g</b>) RPA-CRISPR/Cas12a-PLFS assay for <span class="html-italic">S. terrestris</span>. (<b>e</b>) Optimization of LF-DNA reporter molecule concentrations. 1–5: 50 nmol/L, 100 nmol/L, 200 nmol/L, 400 nmol/L, 800 nmol/L; 1N–5N: ddH<sub>2</sub>O. (<b>f</b>) Optimization of reaction time for visual detection. 1–5: 5 min, 10 min, 15 min, 20 min, 30 min; 1N–5N: ddH<sub>2</sub>O. (<b>g</b>) Sensitivity verification of RPA-CRISPR/Cas12a-PLFS visual detection system. 1–6: 1 ng, 0.1 ng, 10 pg, 1 pg, 0.1 pg, 0.01 pg. N: ddH<sub>2</sub>O.</p>
Full article ">Figure 4
<p>(<b>a</b>–<b>d</b>) Detection of plant samples using RPA-CRISPR/Cas12a assay. (<b>a</b>) RPA detection assay. (<b>b</b>) RPA-CRISPR/Cas12a-FRB. (<b>c</b>) Real-time fluorescent signals in RPA-CRISPR/Cas12a-FRB assay. (<b>d</b>) RPA-CRISPR/Cas12a-PLFS. 1–5: healthy onion samples, sample No. YCCK1, YCCK2, YCCK4, YCCK6, YCCK13, respectively; 6–17: inoculated onion samples with <span class="html-italic">S. terrestris</span>, sample No. YC4, YC6, YC8, YC9, YC11, YC13, YC14, YC15, YC17, YC18, YC19, YC20, respectively; 18: <span class="html-italic">S. terrestris</span>; N: ddH<sub>2</sub>O.</p>
Full article ">
27 pages, 2386 KiB  
Review
Detection Methods for Pine Wilt Disease: A Comprehensive Review
by Sana Tahir, Syed Shaheer Hassan, Lu Yang, Miaomiao Ma and Chenghao Li
Plants 2024, 13(20), 2876; https://doi.org/10.3390/plants13202876 - 14 Oct 2024
Viewed by 603
Abstract
Pine wilt disease (PWD), caused by the nematode Bursaphelenchus xylophilus, is a highly destructive forest disease that necessitates rapid and precise identification for effective management and control. This study evaluates various detection methods for PWD, including morphological diagnosis, molecular techniques, and remote [...] Read more.
Pine wilt disease (PWD), caused by the nematode Bursaphelenchus xylophilus, is a highly destructive forest disease that necessitates rapid and precise identification for effective management and control. This study evaluates various detection methods for PWD, including morphological diagnosis, molecular techniques, and remote sensing. While traditional methods are economical, they are limited by their inability to detect subtle or early changes and require considerable time and expertise. To overcome these challenges, this study emphasizes advanced molecular approaches such as real-time polymerase chain reaction (RT-PCR), droplet digital PCR (ddPCR), and loop-mediated isothermal amplification (LAMP) coupled with CRISPR/Cas12a, which offer fast and accurate pathogen detection. Additionally, DNA barcoding and microarrays facilitate species identification, and proteomics can provide insights into infection-specific protein signatures. The study also highlights remote sensing technologies, including satellite imagery and unmanned aerial vehicle (UAV)-based hyperspectral analysis, for their capability to monitor PWD by detecting asymptomatic diseases through changes in the spectral signatures of trees. Future research should focus on combining traditional and innovative techniques, refining visual inspection processes, developing rapid and portable diagnostic tools for field application, and exploring the potential of volatile organic compound analysis and machine learning algorithms for early disease detection. Integrating diverse methods and adopting innovative technologies are crucial to effectively control this lethal forest disease. Full article
(This article belongs to the Special Issue Biotechnology and Genetic Engineering in Forest Trees)
Show Figures

Figure 1

Figure 1
<p>PWN undergoes a multistage lifecycle, commencing as an egg and progressing through four distinct larval phases (L1 to L4) before ultimately maturing into an adult. Under optimal environmental conditions, these nematodes possess the capability to complete their lifecycle in as few as 4 to 5 days. They rapidly spread to new host trees via their primary vector, the <span class="html-italic">Monochamus</span> beetle, and reproduce within the host tree while primarily feeding on its vascular tissues.</p>
Full article ">Figure 2
<p>Disease progression typically involves a series of stages that delineate the advancement and worsening of a condition over time. In the context of infectious diseases, these stages include the incubation, prodromal, acute, and convalescence periods. Each stage is characterized by distinct symptoms and physiological alterations that influence the strategies employed for treatment and management.</p>
Full article ">Figure 3
<p>The techniques employed for nematode protein identification have the potential to significantly enhance the accuracy and efficiency of diagnostic procedures for species such as <span class="html-italic">B. xylophilus</span> and <span class="html-italic">B. mucronatus</span>. These advancements could substantially impact pest management strategies and ecological research. However, the labor-intensive nature of these techniques and the necessity for a positive reference sample may limit their practical application in rapid field assessments and routine monitoring.</p>
Full article ">Figure 4
<p>Research on <span class="html-italic">B. xylophilus</span> has focused on characterizing its secretome and identifying potential indicators of virulence. Comparative analyses of secretomes and proteomes across multiple <span class="html-italic">B. xylophilus</span> isolates reveal variations in protein expression patterns, which may contribute to differences in nematode pathogenicity and host specificity.</p>
Full article ">Figure 5
<p>Satellite-, aircraft-, or ground-based sensors capture and record reflected or emitted energy across multiple wavelengths of the electromagnetic spectrum to remotely sense an object or area. These remote sensing techniques detect surface features, vegetation health, soil moisture, and other critical properties. The data obtained through these methods is invaluable for applications such as land use mapping, environmental monitoring, and natural resource management.</p>
Full article ">Figure 6
<p>Endophytic fungi and plants collaborate through a biological pathway initiated by plant receptors detecting fungal signals. This recognition triggers a cascade of defense responses and metabolic alterations that mutually benefit both organisms, including enhanced nutrient acquisition and pathogen resistance. During this interaction, metabolites are exchanged, and gene expression is modulated in both partners, facilitating the establishment of a stable symbiotic relationship.</p>
Full article ">Figure 7
<p>This study aims to enhance early detection methods, streamline management strategies, and mitigate the global impact of PWD on pine forest ecosystems.</p>
Full article ">
14 pages, 992 KiB  
Review
Potential Therapeutic Targets for the Treatment of HPV-Associated Malignancies
by Ziyao Lu, Shahab Haghollahi and Muhammad Afzal
Cancers 2024, 16(20), 3474; https://doi.org/10.3390/cancers16203474 - 14 Oct 2024
Viewed by 556
Abstract
This review article aims to summarize broadly recent developments in the treatment of HPV-associated cancers, including cervical cancer and head and neck squamous cell carcinoma. Relatively new treatments targeting the key HPV E6 and E7 oncoproteins, including gene editing with TALENs and CRISPR/Cas9, [...] Read more.
This review article aims to summarize broadly recent developments in the treatment of HPV-associated cancers, including cervical cancer and head and neck squamous cell carcinoma. Relatively new treatments targeting the key HPV E6 and E7 oncoproteins, including gene editing with TALENs and CRISPR/Cas9, are discussed. Given the increased immunogenicity of HPV-related diseases, other therapies such as PRR agonists, adoptive cell transfer, and tumor vaccines are reaching the clinical trial phase. Due to the mechanism, immunogenicity, and reversibility of HPV carcinogenesis, HPV-related cancers present unique targets for current and future therapies. Full article
Show Figures

Figure 1

Figure 1
<p>The role of E6 and E7 in loss of cell cycle control. <span class="html-italic">LCR</span>—Long control region. <span class="html-italic">E6AP</span>—E6 associated protein.</p>
Full article ">
26 pages, 3300 KiB  
Review
Reporter Alleles in hiPSCs: Visual Cues on Development and Disease
by Gustavo Caldeira Cotta, Rachel Castro Teixeira dos Santos, Guilherme Mattos Jardim Costa and Samyra Maria dos Santos Nassif Lacerda
Int. J. Mol. Sci. 2024, 25(20), 11009; https://doi.org/10.3390/ijms252011009 - 13 Oct 2024
Viewed by 505
Abstract
Reporter alleles are essential for advancing research with human induced pluripotent stem cells (hiPSCs), notably in developmental biology and disease modeling. This study investigates the state-of-the-art gene-editing techniques tailored for generating reporter alleles in hiPSCs, emphasizing their effectiveness in investigating cellular dynamics and [...] Read more.
Reporter alleles are essential for advancing research with human induced pluripotent stem cells (hiPSCs), notably in developmental biology and disease modeling. This study investigates the state-of-the-art gene-editing techniques tailored for generating reporter alleles in hiPSCs, emphasizing their effectiveness in investigating cellular dynamics and disease mechanisms. Various methodologies, including the application of CRISPR/Cas9 technology, are discussed for accurately integrating reporter genes into the specific genomic loci. The synthesis of findings from the studies utilizing these reporter alleles reveals insights into developmental processes, genetic disorder modeling, and therapeutic screening, consolidating the existing knowledge. These hiPSC-derived models demonstrate remarkable versatility in replicating human diseases and evaluating drug efficacy, thereby accelerating translational research. Furthermore, this review addresses challenges and future directions in refining the reporter allele design and application to bolster their reliability and relevance in biomedical research. Overall, this investigation offers a comprehensive perspective on the methodologies, applications, and implications of reporter alleles in hiPSC-based studies, underscoring their essential role in advancing both fundamental scientific understanding and clinical practice. Full article
(This article belongs to the Special Issue hiPSC-Based Disease Models as Replacements of Animal Models)
Show Figures

Figure 1

Figure 1
<p>Acquisition and applications of pluripotent stem cells (PSCs). Pluripotent stem cells can be acquired through somatic reprogramming of animal or human cells, such as fibroblasts, or by collecting cells from the inner cell mass of blastocysts. Under very specific culture conditions, their pluripotent potential is maintained, enabling their use in differentiating into lineages of the three germ layers in vitro. Human PSC-derived cells have various applications, including developmental biology studies, disease modeling, organoid development, and cell therapies.</p>
Full article ">Figure 2
<p>Summary of the application of fluorescent reporter alleles for the detection of specific genes (markers) using cellular imaging techniques.</p>
Full article ">Figure 3
<p>Overview of the three major gene-editing platforms. ZFNs, TALENs, and CRISPR/Cas9 rely on the HDR pathway.</p>
Full article ">Figure 4
<p>Experimental workflow required for the generation of iPSCs harboring reporter alleles.</p>
Full article ">
12 pages, 5061 KiB  
Communication
A De Novo Splicing Mutation of STXBP1 in Epileptic Encephalopathy Associated with Hypomyelinating Leukodystrophy
by Zixuan Wang, Jun Zhang, Yunfei Zhou, Guicen Liu, Zixin Tian and Xi Song
Int. J. Mol. Sci. 2024, 25(20), 10983; https://doi.org/10.3390/ijms252010983 - 12 Oct 2024
Viewed by 280
Abstract
Deleterious variations in STXBP1 are responsible for early infantile epileptic encephalopathy type 4 (EIEE4, OMIM # 612164) because of its dysfunction in the central nervous system. The clinical spectrum of the neurodevelopmental delays associated with STXBP1 aberrations is collectively defined as STXBP1 encephalopathy [...] Read more.
Deleterious variations in STXBP1 are responsible for early infantile epileptic encephalopathy type 4 (EIEE4, OMIM # 612164) because of its dysfunction in the central nervous system. The clinical spectrum of the neurodevelopmental delays associated with STXBP1 aberrations is collectively defined as STXBP1 encephalopathy (STXBP1-E), the conspicuous features of which are highlighted by early-onset epileptic seizures without structural brain anomalies. A girl was first diagnosed with unexplained disorders of movement and cognition, which later developed into STXBP1-E with unexpected leukoaraiosis and late onset of seizures. Genetic screening and molecular tests alongside neurological examinations were employed to investigate the genetic etiology and establish the diagnosis. A heterozygous mutation of c.37+2dupT at the STXBP1 splice site was identified as the pathogenic cause in the affected girl. The de novo mutation (DNM) did not result in any truncated proteins but immediately triggered mRNA degradation by nonsense-mediated mRNA decay (NMD), which led to the haploinsufficiency of STXBP1. The patient showed atypical phenotypes characterized by hypomyelinating leukodystrophy, and late onset of epileptic seizures, which had never previously been delineated in STXBP1-E. These findings strongly indicated that the haploinsufficiency of STXBP1 could also exhibit divergent clinical phenotypes because of the genetic heterogeneity in the subset of encephalopathies. Full article
(This article belongs to the Special Issue Exploring Rare Diseases: Genetic, Genomic and Metabolomic Advances)
Show Figures

Figure 1

Figure 1
<p>The course of the disease and clinical features. (<b>A</b>). Course of the disease. A schematic illustration of the clinical records of the proband from her first visit is presented here. (<b>B</b>). Normal EEG. The normal EGG of the girl was recorded at three years old. All * refer to separator. (<b>C</b>). Abnormal EEG. Abnormal EGG of the affected girl was first observed at six years old. The EEG was characterized by focal epileptic activity, burst suppression, hypsarrhythmia, or generalized spike-and-slow waves.</p>
Full article ">Figure 2
<p>Identification of a pathogenic splice variant of c.37+2T in <span class="html-italic">STXBP1</span>. (<b>A</b>) The candidate pathogenic variants screened by trio WES. (<b>B</b>) Confirmation of the variant by direct Sanger sequencing. (<b>C</b>) The pedigree of the family. According to the results, the affected girl harbored the de novo mutation. Arrow marked the proband. (<b>D</b>) The multiple alignments of the adjacent amino acid residues to the donor splice site. The adjacent amino acids were highly conserved between species. Red box highlight the amino acid of the mutation site.</p>
Full article ">Figure 3
<p>The primary cultured neuron cells were bioengineered by CRISPR/Cas9 technology. (<b>A</b>) Confirmation of the bioengineered cell line with c.37+2dupT homogonous mutation in <span class="html-italic">STXBP1</span>. (<b>B</b>) <span class="html-italic">STXBP1</span> mRNA expression detected by RT-PCR. (<b>C</b>) The quantification of the <span class="html-italic">STXBP1</span> mRNA level by q-PCR. The data were presented as mean ± standard deviation (SD). (<b>D</b>) The STXBP1 protein probed by Western blotting.</p>
Full article ">Figure 4
<p>Leukoaraiosis with mild dysmyelination observed in bilateral parietal lobes. Brain MRI from the proband showed diffuse white matter hyperintensity on T2-weighted images (indicated by yellow circles), and the T1-weighted signal represented isointensity, which was consistent with a hypomyelinating leukodystrophy.</p>
Full article ">Figure 5
<p>A therapeutic strategy for splice editing involves two key components: a bioengineered RNA endonuclease and exonuclease, both bearing target antisense oligonucleotides. The endonuclease targets the junction between exon 1 and intron 1, breaking the 3,5 phosphate diester bond at the 3′ terminus of the mutant GUU. Then, the RNA exonuclease removes the duplicated Uridine from the target sequence in exon 1. Endogenous RNA editing and repairing enzymes are then able to rejoin the cleaved <span class="html-italic">STXBP1</span> pre-mRNA. This approach shows promise for therapeutically correcting splice site mutations. Therapeutic editing in pre-mRNA can share this strategy in correcting different mutations, including missense, deletion, and insertion, with dependence on relevant bioengineered RNA repairing partners. Red bubbles represent ssRNA 3′ exonuclease, yellow bubbles represent RNA endonuclease, blue bubbles represent RNA ligase, green bar represent exon1 of <span class="html-italic">STXBP1</span> pre-mRNA and gray bar represent intron1 of <span class="html-italic">STXBP1</span> pre-mRNA.</p>
Full article ">
16 pages, 5040 KiB  
Article
Crispr-SGRU: Prediction of CRISPR/Cas9 Off-Target Activities with Mismatches and Indels Using Stacked BiGRU
by Guishan Zhang, Ye Luo, Huanzeng Xie and Zhiming Dai
Int. J. Mol. Sci. 2024, 25(20), 10945; https://doi.org/10.3390/ijms252010945 - 11 Oct 2024
Viewed by 390
Abstract
CRISPR/Cas9 is a popular genome editing technology, yet its clinical application is hindered by off-target effects. Many deep learning-based methods are available for off-target prediction. However, few can predict off-target activities with insertions or deletions (indels) between single guide RNA and DNA sequence [...] Read more.
CRISPR/Cas9 is a popular genome editing technology, yet its clinical application is hindered by off-target effects. Many deep learning-based methods are available for off-target prediction. However, few can predict off-target activities with insertions or deletions (indels) between single guide RNA and DNA sequence pairs. Additionally, the analysis of off-target data is challenged due to a data imbalance issue. Moreover, the prediction accuracy and interpretability remain to be improved. Here, we introduce a deep learning-based framework, named Crispr-SGRU, to predict off-target activities with mismatches and indels. This model is based on Inception and stacked BiGRU. It adopts a dice loss function to solve the inherent imbalance issue. Experimental results show our model outperforms existing methods for off-target prediction in terms of accuracy and robustness. Finally, we study the interpretability of this model through Deep SHAP and teacher–student-based knowledge distillation, and find it can provide meaningful explanations for sequence patterns regarding off-target activity. Full article
Show Figures

Figure 1

Figure 1
<p>Heatmaps of precision, recall, F1 score, and MCC values of Crispr-SGRU and four existing methods on eight datasets with varying imbalance ratios. The predictors are placed vertically, whereas the test datasets are arranged horizontally. Datasets are sorted by imbalance ratio in ascending order.</p>
Full article ">Figure 2
<p>ROC plots for Crispr-SGRU and four deep learning-based methods on eight datasets under five-fold cross-validation.</p>
Full article ">Figure 3
<p>PR plots for Crispr-SGRU and four deep learning-based methods on eight datasets under five-fold cross-validation.</p>
Full article ">Figure 4
<p>Performance comparison of Crispr-SGRU with four other methods (e.g., CRISPR-Net, CRISPR-IP, CrisprDNT, and CRISPR-M) on datasets K562, HEK293t, BE3, and II5 under a leave-one-sgRNA-out test.</p>
Full article ">Figure 5
<p>ROC and PR plots of Crispr-SGRU with four predictors (CRISPR-Net, CRISPR-IP, CrisprDNT, and CRISPR-M) implemented on four datasets—K562, HEK293t, BE3, and II5—under a leave-one-sgRNA-out test.</p>
Full article ">Figure 6
<p>Visualization of the importance of various positions for Crispr-SGRU on six mismatch-only datasets. The color of each cell in the heatmap represents the contribution of nucleotide positions to the off-target prediction.</p>
Full article ">Figure 7
<p>Visualization of importance of the sequence context for each position in sgRNA–DNA sequence pairs for Student with KD model on K562 dataset. The nucleotide positions are arranged horizontally, whereas various sequence contexts are placed vertically. Color and size of the dots in the bubble plot represent the average Deep SHAP values, which represent the contribution of nucleotide position to off-target prediction.</p>
Full article ">Figure 8
<p>(<b>a</b>) The structure of Crispr-SGRU. Each sgRNA–DNA sequence pair is encoded and fed into an Inception module (<b>b</b>) for feature extraction. The outputs of Inception are put into the stacked BiGRU (<b>c</b>) to learn the sequential dependencies of the sequence pairs. The number of units in each BiGRU is 30, 20, and 10. The outputs of stacked BiGRU layers are concatenated and passed through three dense layers with 128, 64, and 2 neurons, respectively. A sigmoid activation function is applied in the output layer to obtain the final prediction results.</p>
Full article ">Figure 9
<p>The teacher–student framework for knowledge distillation. (<b>a</b>) Workflow of knowledge distillation. Crispr-SGRU is the teacher model. The student model is applied to increase the interpretability of Crispr-SGRU. (<b>b</b>) Overview of the encoding scheme and (<b>c</b>) student model.</p>
Full article ">
14 pages, 2886 KiB  
Article
DeepIndel: An Interpretable Deep Learning Approach for Predicting CRISPR/Cas9-Mediated Editing Outcomes
by Guishan Zhang, Huanzeng Xie and Xianhua Dai
Int. J. Mol. Sci. 2024, 25(20), 10928; https://doi.org/10.3390/ijms252010928 - 11 Oct 2024
Viewed by 416
Abstract
CRISPR/Cas9 has been applied to edit the genome of various organisms, but our understanding of editing outcomes at specific sites after Cas9-mediated DNA cleavage is still limited. Several deep learning-based methods have been proposed for repair outcome prediction; however, there is still room [...] Read more.
CRISPR/Cas9 has been applied to edit the genome of various organisms, but our understanding of editing outcomes at specific sites after Cas9-mediated DNA cleavage is still limited. Several deep learning-based methods have been proposed for repair outcome prediction; however, there is still room for improvement in terms of performance regarding frameshifts and model interpretability. Here, we present DeepIndel, an end-to-end multi-label regression model for predicting repair outcomes based on the BERT-base module. We demonstrate that our model outperforms existing methods in terms of accuracy and generalizability across various metrics. Furthermore, we utilized Deep SHAP to visualize the importance of nucleotides at various positions for DNA sequence and found that mononucleotides and trinucleotides in DNA sequences surrounding the cut site play a significant role in repair outcome prediction. Full article
(This article belongs to the Special Issue Machine Learning Applications in Bioinformatics and Biomedicine 2.0)
Show Figures

Figure 1

Figure 1
<p>Performance comparison of DeepIndel with three model variants on the K562 dataset under 5-fold cross-validation. The performance comparison in terms of (<b>a</b>) AUC, (<b>b</b>) SCC, (<b>c</b>) PCC, and (<b>d</b>) KTau. The prediction methods are arranged vertically, whereas the repair outcomes are arranged horizontally. DelF, deletion frequency; 1 InsF, 1 bp insertion frequency; 1 DelF, 1 bp deletion frequency; 1 FsF, 1 bp frameshift frequency; 2 FsF, 2 bp frameshift frequency; FsF, total frameshift frequency, and the average of all prediction tasks. These representations also apply to Figures 2–5 and <a href="#app1-ijms-25-10928" class="html-app">Supplementary Figures S1–S5</a>. Error bars represent the standard deviation.</p>
Full article ">Figure 2
<p>The bar graphs show averaged (<b>a</b>) AUC, (<b>b</b>) SCC, (<b>c</b>) PCC, and (<b>d</b>) KTau values of DeepIndel and two deep learning-based methods on the K562 dataset under 5-fold cross-validation. The Error bars represent the standard deviation.</p>
Full article ">Figure 3
<p>The bar graphs show the (<b>a</b>) AUC, (<b>b</b>) SCC, (<b>c</b>) PCC, and (<b>d</b>) KTau of DeepIndel with two existing deep learning-based methods (e.g., Apindel and CROTON) under cross-dataset validation tests. The models were trained on the K562 dataset and their performance was tested on the T cell dataset. The error bars represent the standard deviation. The dotted line in (<b>a</b>) indicates an AUC of 0.5.</p>
Full article ">Figure 4
<p>Visualization of the feature importance of 1-mer, 2-mer, and 3-mer sequences at each position learned by DeepIndel on the (<b>a</b>) K562, (<b>b</b>) HEK293t, and (<b>c</b>) T cell datasets, respectively. A positive value indicates a favored feature, whereas a negative value means a disfavored feature.</p>
Full article ">Figure 5
<p>Visualization of the feature importance of nucleotide position in the target DNA sequence estimated by DeepIndel for six repair outcomes on the (<b>a</b>) K562, (<b>b</b>) HEK293t, and (<b>c</b>) T cell datasets. The nucleotide positions are arranged horizontally, whereas the datasets are arranged vertically. The color of each cell in the heatmap denotes the contribution of trinucleotides at each position for repair outcome prediction.</p>
Full article ">Figure 6
<p>Sequence encoding for denoting the target DNA sequence. (<b>a</b>) Rules of setting index values for two special symbols (e.g., [CLS] and [SEP]), 4 single nucleotides, 16 dinucleotides, and 64 trinucleotides. (<b>b</b>) An example showing how to use the proposed token dictionary to encode the DNA sequence.</p>
Full article ">Figure 7
<p>Illustration of DeepIndel and the transformer block in BERT. DeepIndel predicts the repair results by the following six stages. (i) A 60 bp DNA sequence centered at the cut site is used as the input to the model. The encoding layer converts the input sequence into a numerical sequence of length 179. (ii) The encoded sequence is fed into BERT to extract the hidden information. (iii) The outputs of BERT are input into the first dense layer for deeper feature extraction with the ReLU activation function. (iv) The outputs of the dense layer are flattened for further analysis. (v) The features are fed into the second dense layer to perform linear transformations. (vi) The output layer makes a multi-output regression prediction of repair outcomes.</p>
Full article ">
30 pages, 14754 KiB  
Review
Recent Advances in the CRISPR/Cas-Based Nucleic Acid Biosensor for Food Analysis: A Review
by Yanan Sun, Tianjian Wen, Ping Zhang, Minglian Wang and Yuancong Xu
Foods 2024, 13(20), 3222; https://doi.org/10.3390/foods13203222 - 10 Oct 2024
Viewed by 778
Abstract
Food safety is a major public health issue of global concern. In recent years, the CRISPR/Cas system has shown promise in the field of molecular detection. The system has been coupled with various nucleic acid amplification methods and combined with different signal output [...] Read more.
Food safety is a major public health issue of global concern. In recent years, the CRISPR/Cas system has shown promise in the field of molecular detection. The system has been coupled with various nucleic acid amplification methods and combined with different signal output systems to develop a new generation of CRISPR/Cas-based nucleic acid biosensor technology. This review describes the design concept of the CRISPR/Cas-based nucleic acid biosensor and its application in food analysis. A detailed overview of different CRISPR/Cas systems, signal amplification methods, and signal output strategies is provided. CRISPR/Cas-based nucleic acid biosensors have the advantages of high sensitivity, strong specificity, and timeliness, achieving fast analysis of a variety of targets, including bacteria, toxins, metal ions, pesticides, veterinary drugs, and adulteration, promoting the development of rapid food safety detection technology. At the end, we also provide our outlook for the future development of CRISPR/Cas-based nucleic acid biosensors. Full article
(This article belongs to the Special Issue Recent Advances in Biosensor Applications for Food Products)
Show Figures

Figure 1

Figure 1
<p>The CRISPR/Cas-based nucleic acid biosensor of food analysis. The blue part indicates nucleic acid amplification methods. The orange part indicates signal output analytical systems. The green part indicates food analysis targets.</p>
Full article ">Figure 2
<p>The principal diagram of signal amplification and signal output in the CRISPR/Cas-based nucleic acid biosensors. (<b>A</b>) Schematic illustration of fluorescence signal without nucleic acid amplification. Reprinted with permission from [<a href="#B35-foods-13-03222" class="html-bibr">35</a>]. Copyright 2019 American Chemical Society. (<b>B</b>) Schematic illustration of LAMP combined with lateral flow strips [<a href="#B40-foods-13-03222" class="html-bibr">40</a>]. Copyright 2020 Elsevier B.V. (<b>C</b>) Schematic illustration of PCR/RPA combined with AuNPs colorimetry. Reprinted with permission from [<a href="#B42-foods-13-03222" class="html-bibr">42</a>]. Copyright 2022 Elsevier Ltd. (<b>D</b>) Schematic illustration of RCA combined with electrochemical signals. Reprinted with permission from [<a href="#B43-foods-13-03222" class="html-bibr">43</a>]. Copyright 2021 American Chemical Society. (<b>E</b>) Schematic illustration of HCR combined with G4 colorimetry. Reprinted with permission from [<a href="#B44-foods-13-03222" class="html-bibr">44</a>]. Copyright 2023 American Chemical Society. (<b>F</b>) Schematic illustration of SERS signal without nucleic acid amplification. Reprinted with permission from [<a href="#B45-foods-13-03222" class="html-bibr">45</a>]. Copyright 2023 Elsevier B.V.</p>
Full article ">Figure 3
<p>Principal diagram of foodborne pathogen detection by CRISPR/Cas-based nucleic acid biosensors. (<b>A</b>) Schematic illustration of the label-free colorimetric biosensor for the detection of <span class="html-italic">Vibrio parahaemolyticus</span>. In the presence of <span class="html-italic">Vibrio parahaemolyticus</span>, LAMP products combine with crRNA. The active Cas12a cleaves the G4 probe and is unable to catalyze a color change in ABTS<sup>2−</sup>. The positive and negative results can then be easily distinguished by the obvious color differences. Reprinted with permission from [<a href="#B87-foods-13-03222" class="html-bibr">87</a>]. Copyright 2021 American Chemical Society. (<b>B</b>) Schematic illustration of the CATCHER for the detection of <span class="html-italic">Salmonella Typhimurium</span>. Once the “MNP-Ab/<span class="html-italic">S. Typhimurium</span>/CG-Ab-DNase I” is formed, DNase I converts activator DNA to invalid DNA fragments, resulting in the inactivation of Cas12a and the silencing of the signal output system. Reprinted with permission from [<a href="#B90-foods-13-03222" class="html-bibr">90</a>]. Copyright 2022 Elsevier B.V.</p>
Full article ">Figure 4
<p>Principal diagram of biotech food detection by CRISPR/Cas-based nucleic acid biosensors. (<b>A</b>) Schematic illustration of Cas12a-PB for the detection of GM soybeans. LAMP assay reagents and CRISPR/Cas12a assay reagents were preloaded onto PCR tubes and detection chambers of PMMA, respectively. After amplification, the reaction tube was inverted and the detection was completed by fluorescence changes in different detection chambers. Reprinted with permission from [<a href="#B95-foods-13-03222" class="html-bibr">95</a>]. Copyright 2020 Elsevier B.V. (<b>B</b>) Schematic illustration of the SERRS assay for the identification of GMOs. A magnetic SERRS nanoprobe (FeAuG-MB) was constructed by modifying the G-quadruplex on the surface of gold-coated magnetic beads. Methylene blue (MB) was inserted into the G-quadruplex as a SERS reporter molecule. When the <span class="html-italic">trans</span>-cleavage of the CRISPR/Cas12a was activated, the G-triplet DNA was cleaved, which caused a release of methylene blue above the nanoprobes and a reduction in the SERRS signal. Reprinted with permission from [<a href="#B92-foods-13-03222" class="html-bibr">92</a>]. Copyright 2024 Elsevier B.V.</p>
Full article ">Figure 5
<p>Principal diagram of food adulteration detection by CRISPR/Cas-based nucleic acid biosensors. (<b>A</b>) Schematic illustration of the ECL biosensor for the detection of pufferfish. This involved NiCo<sub>2</sub>O<sub>4</sub> NCs@Au-ABEI designed as nanoemitters that were immobilized on the electrode surface by nucleic acid hybridization. The <span class="html-italic">trans</span>-cleavage activity of CRISPR/Cas12a was initiated when pufferfish DNA was present, leading to a nonspecifically cleaved ssDNA and releasing NiCo<sub>2</sub>O<sub>4</sub> NCs@Au-ABEI. As a result, the ECL signal was significantly increased. Reprinted with permission from [<a href="#B98-foods-13-03222" class="html-bibr">98</a>]. Copyright 2024 Elsevier Ltd. (<b>B</b>) Schematic illustration of CIPAM for the simultaneous detection of pig, chicken, duck, and lamb. The modules of the centrifugal microfluidic chip were pre-loaded with the reagents required for the RAA-CRISPR/Cas12a reaction for integrated operation. If target DNA was present, a fluorescent signal appeared in the corresponding reaction module. Reprinted with permission from [<a href="#B100-foods-13-03222" class="html-bibr">100</a>]. Copyright 2024 Elsevier Ltd.</p>
Full article ">Figure 6
<p>Principal diagram of biotoxin detection by CRISPR/Cas-based nucleic acid biosensors. (<b>A</b>) Schematic illustration of the neoteric electrochemical biosensor for the detection of OTA. Using an Exo III-assisted recirculation reaction, the target recognition transaction was converted into a triggering DNA strand and amplified into the <span class="html-italic">trans</span>-cleavage activity of Cas12a to detect changes in electrochemical signals. Reprinted with permission from [<a href="#B103-foods-13-03222" class="html-bibr">103</a>]. Copyright 2023 Elsevier B.V. (<b>B</b>) Schematic illustration of the hydrazone ligation-assisted DNAzyme walking nanomachine for the detection of lipopolysaccharide. In the presence of LPS, LPS bound to the aptamer in the complex to release ASII, triggering subsequent reactions and enhancing fluorescence. Reprinted with permission from [<a href="#B104-foods-13-03222" class="html-bibr">104</a>]. Copyright 2021 Elsevier B.V.</p>
Full article ">Figure 7
<p>Principal diagram of heavy metal ion detection by CRISPR/Cas-based nucleic acid biosensors. (<b>A</b>) Schematic illustration of the dual-functional DNAzyme-powered CRISPR-Cas12a biosensor for the detection of Pb<sup>2+</sup>. In the presence of Pb<sup>2+</sup>, the DNAzyme was cleaved, causing disintegration of the MB-DNAzyme/AuNP probe. The degraded DNAzyme/AuNP in the supernatant effectively triggered CRISPR/Cas12a activity, initiating DNA reporter cleavage and generating great fluorescence. Reprinted with permission from [<a href="#B108-foods-13-03222" class="html-bibr">108</a>]. Copyright 2023 Elsevier B.V. (<b>B</b>) Schematic illustration of the paper-based microfluidic biosensor for the detection of Pb<sup>2+</sup>. The Pb<sup>2+</sup>-induced G-quadruplex inhibited the Cas12a activity, which, in turn, prevented the cleavage of the ssDNA probe around AuNPs. Subsequently, ssDNA-AuNPs flowed into the paper-based microfluidic chip and were captured by the immobilized nanoflowers, resulting in a corresponding color change. Reprinted with permission from [<a href="#B110-foods-13-03222" class="html-bibr">110</a>]. Copyright 2023 Elsevier B.V.</p>
Full article ">Figure 8
<p>Principal diagram of pesticide and veterinary drug detection by CRISPR/Cas-based nucleic acid biosensors. (<b>A</b>) Schematic illustration of TCApt-EDC-CHA-Cas12a sensing for the detection of tetracycline. In the presence of tetracycline, the release of the complementary strand from the aptamer triggered the cascade amplification circuit, which, in turn, activated the activity of Cas12a to cleave FAM-ssDNA to restore fluorescence. Reprinted with permission from [<a href="#B117-foods-13-03222" class="html-bibr">117</a>]. Copyright 2023 Wiley-VCH GmbH. (<b>B</b>) Schematic illustration of the AChE-mediated fluorescence biosensor for the detection of OPs. In the absence of OPs, TCh containing reducing sulfhydryl groups was generated, which induced the degradation of MnO<sub>2</sub> nanosheets and produced Mn<sup>2+</sup> to cleavage the Mn<sup>2+</sup>-dependent DNAzyme. Then, the released short DNA activated the activity of Cas12a to cleavage ssDNA probes, resulting in a change in fluorescence. Reprinted with permission from [<a href="#B114-foods-13-03222" class="html-bibr">114</a>]. Copyright 2022 Elsevier Ltd.</p>
Full article ">
15 pages, 3428 KiB  
Article
ddPCR Overcomes the CRISPR-Cas13a-Based Technique for the Detection of the BRAF p.V600E Mutation in Liquid Biopsies
by Irina Palacín-Aliana, Noemí García-Romero, Josefa Carrión-Navarro, Pilar Puig-Serra, Raul Torres-Ruiz, Sandra Rodríguez-Perales, David Viñal, Víctor González-Rumayor and Ángel Ayuso-Sacido
Int. J. Mol. Sci. 2024, 25(20), 10902; https://doi.org/10.3390/ijms252010902 - 10 Oct 2024
Viewed by 365
Abstract
The isolation of circulating tumoral DNA (ctDNA) present in the bloodstream brings about the opportunity to detect genomic aberrations from the tumor of origin. However, the low amounts of ctDNA present in liquid biopsy samples makes the development of highly sensitive techniques necessary [...] Read more.
The isolation of circulating tumoral DNA (ctDNA) present in the bloodstream brings about the opportunity to detect genomic aberrations from the tumor of origin. However, the low amounts of ctDNA present in liquid biopsy samples makes the development of highly sensitive techniques necessary to detect targetable mutations for the diagnosis, prognosis, and monitoring of cancer patients. Here, we employ standard genomic DNA (gDNA) and eight liquid biopsy samples from different cancer patients to examine the newly described CRISPR-Cas13a-based technology in the detection of the BRAF p.V600E actionable point mutation and appraise its diagnostic capacity with two PCR-based techniques: quantitative Real-Time PCR (qPCR) and droplet digital PCR (ddPCR). Regardless of its lower specificity compared to the qPCR and ddPCR techniques, the CRISPR-Cas13a-guided complex was able to detect inputs as low as 10 pM. Even though the PCR-based techniques have similar target limits of detection (LoDs), only the ddPCR achieved a 0.1% variant allele frequency (VAF) detection with elevated reproducibility, thus standing out as the most powerful and suitable tool for clinical diagnosis purposes. Our results also demonstrate how the CRISPR-Cas13a can detect low amounts of the target of interest, but its base-pair specificity failed in the detection of actionable point mutations at a low VAF; therefore, the ddPCR is still the most powerful and suitable technique for these purposes. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of nucleic acid mutation detection using CRISPR-Cas13a enzyme collateral cleavage activity. (<b>A</b>) Schematic image of sample extraction and steps needed for the ssRNA acquisition. After genomic DNA (gDNA) or circulating free DNA (cfDNA) extraction from the clinical samples, the first step consists of DNA amplification by conventional PCR performed with primers tagged with a T7 promoter sequence. The pre-amplification step will generate double-stranded DNA (dsDNA) amplicons of the target sequence with an appended T7 promoter sequence needed for the next step and for procedure sensibility improvement. Thereafter, the in vitro transcription (IVT) of the PCR product by a T7 polymerase will produce transcribed single-stranded RNA (ssRNA) targets. (<b>B</b>) Representation of the collateral cleavage Cas13a: crRNA complex activated by ssRNA target sequence binding. The CRISPR guide sequence (crRNA) contains repeat sequences that will form a loop essential for its anchor to the Cas13a nuclease. Once the Cas13a: crRNA complex has been formed, the crRNA and ssRNA target base-pairing activates the collateral nuclease activity of the Cas13a. This collateral cleavage activity will cleave a fluorescent reporter attached to its quencher by a short ssRNA sequence generating a measurable fluorescent signal. (<b>C</b>) Schematic illustration of the Cas13a: crRNA complex. Two different crRNA guide sequences have been designed for the detection of the <span class="html-italic">BRAF</span> wild-type (WT) and <span class="html-italic">BRAF</span> p.V600E-mutated sequences. The Cas13a: crRNA complex is formed by the loop sequence present in the crRNA. Cas13a is inactive when it is not bound to target ssRNA. Once the complex binds to the ssRNA, the collateral RNAse activity of the Cas13a is initiated. To enhance the single-base pair specificity, an additional synthetic mismatch is placed next to the point mutation of interest (marked in red).</p>
Full article ">Figure 2
<p>CRISPR-Cas13a ssRNA target detection. (<b>A</b>) Fluorescent measurement of Cas13a activity employing 50 nM of ssRNA target. RNase A was used as positive control for the cleavage of the fluorescent RNA reporter. (<b>B</b>) CRISPR-Cas13a time-course fluorescence signal intensities expressed in logarithmic scale under different ssRNA target concentration inputs and using the BRAF p.V600E crRNA (10 pM, 50 pM, 100 pM, 500 pM, 1 nM, 10 nM, 50 nM, and 250 nM). Fluorescence measurements were taken every 5 min at 37 °C. (<b>C</b>,<b>D</b>) Linear relationship between final fluorescent signal (t = 1 h) ((<b>B</b>) data) and ssRNA target concentration (10 pM, 50 pM, 100 pM, 500 pM, 1 nM, 10 nM, 50 nM, and 250 nM). <span class="html-italic">n</span> = 3 independent experimental reactions with technical duplicates; error bars represent mean ± SD.</p>
Full article ">Figure 3
<p>CRISPR-Cas13a <span class="html-italic">BRAF</span> p.V600E mutation detection. CRISPR-Cas13a minor allele frequency detection using different ssRNA target inputs: (<b>A</b>) 250 nM, (<b>B</b>) 10 nM, (<b>C</b>) 1 nM, (<b>D</b>) 500 pM, (<b>E</b>) 100 pM. <span class="html-italic">n</span> = 3 independent experimental duplicates; all readings were made after 1h of reaction incubation; bars represent mean ± SD; two-tailed <span class="html-italic">t</span> test against the WT ssRNA (grey): *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001; ****, <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>qPCR <span class="html-italic">BRAF</span> p.V600E limit of detection characterization. CRISPR-Cas13a minor allele frequency detection using different ssRNA target inputs. (<b>A</b>) qPCR <span class="html-italic">BRAF</span> p.V600E signal amplifications under different inputs of target concentrations (250 to 1 nM). (<b>B</b>) qPCR mutant allele frequency. <span class="html-italic">n</span> = 3 independent experimental assays; bars represent mean ± SD; two-tailed <span class="html-italic">t</span> test.</p>
Full article ">Figure 5
<p>ddPCR <span class="html-italic">BRAF</span> p.V600E limit of detection characterization. CRISPR-Cas13a minor allele frequency detection using different ssRNA target inputs. (<b>A</b>) ddPCR <span class="html-italic">BRAF</span> p.V600E input quantification under different inputs of target concentrations (250 to 1 nM). (<b>B</b>) ddPCR mutant allele frequency detection. (<b>C</b>) Sample fractional abundance. <span class="html-italic">n</span> = 3 independent experimental assays; bars represent mean ± SD; two-tailed <span class="html-italic">t</span> test; ****, <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>Comparison of CRISPR-Cas13a reproducibility to other nucleic acid detection tools. (<b>A</b>) Coefficient of variation of CRISPR-Cas13a minor allele frequency detection using different ssRNA target inputs. (<b>B</b>) Coefficient of variation of qPCR minor allele frequency detection using different target inputs. (<b>C</b>) Coefficient of variation of ddPCR minor allele frequency detection using different target inputs. (<b>D</b>) Mean coefficient of variation for different target inputs and the three detection methods. <span class="html-italic">n</span> = 3 independent experimental duplicates; bars represent mean ± SD; two-tailed <span class="html-italic">t</span> test: ****, <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Detection of <span class="html-italic">BRAF</span> p.V600E from lung and colorectal cancer patients’ cfDNA by CRISPR-Cas13a, qPCR, and ddPCR. (<b>A</b>) CRISPR-Cas13a collateral activity for the detection of <span class="html-italic">BRAF</span> p.V600E alteration. (<b>B</b>) qPCR <span class="html-italic">BRAF</span> p.V600E alteration amplification employing a WT and altered probes. (<b>C</b>) ddPCR <span class="html-italic">BRAF</span> p.V600E alteration quantification employing a WT and altered probes. (<b>D</b>) Sample fractional abundance identified via ddPCR. <span class="html-italic">n</span> = 2 independent experimental duplicates.</p>
Full article ">
13 pages, 2371 KiB  
Article
Rapid and Ultrasensitive Detection of H. aduncum via the RPA-CRISPR/Cas12a Platform
by Xiaoming Wang, Xiang Chen, Ting Xu, Xingsheng Jin, Junfang Jiang and Feng Guan
Molecules 2024, 29(20), 4789; https://doi.org/10.3390/molecules29204789 - 10 Oct 2024
Viewed by 428
Abstract
Hysterothylacium aduncum is one of six pathogens responsible for human anisakiasis. Infection with H. aduncum can cause acute abdominal symptoms and allergic reactions and is prone to misdiagnosis in clinical practice. This study aims to enhance the efficiency and accuracy of detecting H. [...] Read more.
Hysterothylacium aduncum is one of six pathogens responsible for human anisakiasis. Infection with H. aduncum can cause acute abdominal symptoms and allergic reactions and is prone to misdiagnosis in clinical practice. This study aims to enhance the efficiency and accuracy of detecting H. aduncum in food ingredients. We targeted the internal transcribed spacer 1 (ITS 1) regions of Anisakis to develop a visual screening method for detecting H. aduncum using recombinase polymerase amplification (RPA) combined with the CRISPR/Cas12a system. By comparing the ITS 1 region sequences of eight nematode species, we designed specific primers and CRISPR RNA (crRNA). The specificity of RPA primers was screened and evaluated, and the CRISPR system was optimized. We assessed its specificity and sensitivity and performed testing on commercial samples. The results indicated that the alternative primer ADU 1 was the most effective. The final optimized concentrations were 250 nM for Cas12a, 500 nM for crRNA, and 500 nM for ssDNA. The complete test procedure was achievable within 45 min at 37 °C, with a limit of detection (LOD) of 1.27 pg/μL. The amplified product could be directly observed using a fluorescence microscope or ultraviolet lamp. Detection results for 15 Anisakis samples were entirely consistent with those obtained via Sanger sequencing, demonstrating the higher efficacy of this method for detecting and identifying H. aduncum. This visual detection method, characterized by simple operation, visual results, high sensitivity, and specificity, meets the requirements for food safety testing and enhances monitoring efficiency. Full article
(This article belongs to the Section Food Chemistry)
Show Figures

Figure 1

Figure 1
<p>RPA primer optimization results. Lanes 2, 5, and 8 show the amplification results for the genomic primer pairs ADU1, ADU2, and ADU3 of <span class="html-italic">H. aduncum</span>; Lanes 1, 4, and 7 are the negative controls using <span class="html-italic">A. simplex</span> (<span class="html-italic">s. s.</span>); Lanes 3, 6, and 9 are the negative controls using ddH<sub>2</sub>O.</p>
Full article ">Figure 2
<p>Positions of RPA primers and crRNA target sequences for <span class="html-italic">H. aduncum</span>.</p>
Full article ">Figure 3
<p>The results for specificity based on the RPA-CRISPR/Cas12a for <span class="html-italic">H. aduncum</span>. (<b>A</b>) Endpoint fluorescence analysis of the RPA-CRISPR/Cas12a assay. (<b>B</b>) Fluorescent color development under UV light of the RPA-CRISPR/Cas12a assay. (<b>C</b>) Specificity test of the RPA reaction. Lane 1 represents <span class="html-italic">H. aduncum</span>; Lane 2 represents <span class="html-italic">A. simplex</span> (<span class="html-italic">s. s.</span>); Lane 3 represents <span class="html-italic">A. typica</span>; Lane 4 represents <span class="html-italic">H. sinense</span>; Lane 5 represents <span class="html-italic">Contracaecum</span> spp.; Lane 6 represents the aggregate sample of two fish species meat; Lane 7 represents negative control (ddH<sub>2</sub>O was used as a reaction template). ** represent <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Optimization of Cas12a/crRNA concentration ratio. (<b>A</b>) Endpoint fluorescence analysis of the system; (<b>B</b>) Fluorescent color development under UV light. ** represent <span class="html-italic">p</span> &lt; 0.01; ns represent <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 5
<p>Optimization of ssDNA concentration. (<b>A</b>) Endpoint fluorescence analysis of the RPA-CRISPR/Cas12a assay; (<b>B</b>) fluorescent color development under UV light. ** represent <span class="html-italic">p</span> &lt; 0.01; ns represent <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 6
<p>Sensitivity of the RPA-CRISPR/Cas12a system. (<b>A</b>) Sensitivity evaluation of the RPA reaction. (<b>B</b>) Endpoint fluorescence analysis of the RPA-CRISPR/Cas12a assay. (<b>C</b>) Fluorescent color development under UV light of the RPA-CRISPR/Cas12a assay. Lane 1: 12.7 ng/μL; Lane 2: 1.27 ng/μL; Lane 3: 127 pg/μL; Lane 4: 12.7 pg/μL; Lane 5: 1.27 pg/μL; Lane 6: 127 fg/μL; Lane 7: 12.7 fg/μL; Lane 8: ddH<sub>2</sub>O. ** represent <span class="html-italic">p &lt;</span> 0.01; ns represent <span class="html-italic">p &gt;</span> 0.05.</p>
Full article ">Figure 7
<p>Results of actual sample testing using the RPA-CRISPR/Cas12a assay. (<b>A</b>) Endpoint fluorescence analysis of the RPA-CRISPR/Cas12a assay. (<b>B</b>) Fluorescent color development under UV light.</p>
Full article ">
8 pages, 1005 KiB  
Communication
Establishment of a Genetic Transformation and Gene Editing Method by Floral Dipping in Descurainia sophia
by Tianjiao Jia, Hua Yang, Dingding Zhou, Sanzeng Zhao, Jianyong Wang, Tao Zhang, Mingkun Huang, Danyu Kong and Yi Liu
Plants 2024, 13(20), 2833; https://doi.org/10.3390/plants13202833 - 10 Oct 2024
Viewed by 515
Abstract
Descurainia sophia L. Webb ex Prantl is used in traditional medicine globally. However, the lack of an efficient and reliable genetic transformation system has seriously limited the investigation of gene function and further utilization of D. sophia. In this study, a highly [...] Read more.
Descurainia sophia L. Webb ex Prantl is used in traditional medicine globally. However, the lack of an efficient and reliable genetic transformation system has seriously limited the investigation of gene function and further utilization of D. sophia. In this study, a highly efficient, time-saving, and cost-effective Agrobacterium tumefaciens-mediated genetic transformation system has been developed in D. sophia. In this method, the transformation was accomplished by simply dipping developing D. sophia inflorescences for 45 s into an Agrobacterium suspension (OD600 = 0.6) containing 5% sucrose and 0.03% (v/v) Silwet L-77. Treated plants were allowed to set seeds which were then plated on a selective medium with hygromycin B (HygB) to screen transformants. Additionally, the CRISPR/Cas9 genomic editing system was validated by targeting phytoene desaturase (PDS) gene using this floral dip method, and mutant plants with the expected albino phenotype could be obtained in 2.5 months. This genetic transformation and targeted editing system will be a valuable tool for routine investigation of gene function and further exploitation in D. sophia. Full article
(This article belongs to the Special Issue Advances and Applications of Genome Editing in Plants)
Show Figures

Figure 1

Figure 1
<p><b><span class="html-italic">A. tumefaciens</span>-mediated transformation and gene editing of</b> <span class="html-italic">D. sophia</span> <b>with the floral dip method.</b> (<b>a</b>) Schematic of <span class="html-italic">Agrobacterium</span>-mediated transformation of <span class="html-italic">D. sophia</span> by the floral dip method. (<b>b</b>) Transformation efficiencies for <span class="html-italic">D. sophia</span> under different OD<sub>600</sub> values of <span class="html-italic">Agrobacterium</span>, AS concentrations, and Silwet L-77 concentrations. All seeds yielded by each plant have been sown on the selection plates in this assay. Data are mean ± SD. (<b>c</b>) GUS staining analysis of leaf and flower from WT <span class="html-italic">D. sophia</span> plants (1, 3) and T<sub>1</sub> transgenic plants (2, 4). Bar: 400 μm. (<b>d</b>) GFP analysis of WT and transgenic <span class="html-italic">D. sophia</span> plants. The photos of leaf and root from WT <span class="html-italic">D. sophia</span> plant (1, 2) and T<sub>1</sub> transgenic seedling (5, 6) were taken under white light, respectively. The photos of leaf and root from WT <span class="html-italic">D. sophia</span> (3, 4) and T<sub>1</sub> transgenic seedling (7, 8) were taken in fluorescence, respectively. Bar: 100 μm (1, 3, 5, and 7), 50 μm (2, 4, 6, and 8). (<b>e</b>) Representative photo of a WT <span class="html-italic">D. sophia</span> and a T<sub>1</sub> seedling with <span class="html-italic">DsPDS</span> edited. WT seedlings grew on 1/2 MS media, while mutant seedlings were screened from 1/2 MS media containing 50 mg/L of HygB. (<b>f</b>) The structure of <span class="html-italic">DsPDS</span> and the indel efficiency of <span class="html-italic">D. sophia</span> mutants analyzed by ICE. Coding sequences (CDS) of <span class="html-italic">DsPDS</span> are shown by blue boxes. The locations of sgRNA are illustrated by red triangles. (<b>g</b>) The editing profile of plant Sanger sequencing. The contributions show the inferred sequencing present in the edited population and their relative proportion. The cut site is represented by black dotted vertical line. The sgRNA target sequences are labeled by the black lines. The protospacer-adjacent motifs are labeled by red dotted horizontal lines.</p>
Full article ">
Back to TopTop