[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (318)

Search Parameters:
Keywords = agarose gels

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 4256 KiB  
Article
Sulfated and Phosphorylated Agarose as Biomaterials for a Biomimetic Paradigm for FGF-2 Release
by Aurelien Forget and V. Prasad Shastri
Biomimetics 2025, 10(1), 12; https://doi.org/10.3390/biomimetics10010012 - 30 Dec 2024
Viewed by 234
Abstract
Cardiovascular diseases such as myocardial infarction or limb ischemia are characterized by regression of blood vessels. Local delivery of growth factors (GFs) involved in angiogenesis such as fibroblast blast growth factor-2 (FGF-2) has been shown to trigger collateral neovasculature and might lead to [...] Read more.
Cardiovascular diseases such as myocardial infarction or limb ischemia are characterized by regression of blood vessels. Local delivery of growth factors (GFs) involved in angiogenesis such as fibroblast blast growth factor-2 (FGF-2) has been shown to trigger collateral neovasculature and might lead to a therapeutic strategy. In vivo, heparin, a sulfated polysaccharide present in abundance in the extracellular matrix (ECM), has been shown to function as a local reservoir for FGF-2 by binding FGF-2 and other morphogens and it plays a role in the evolution of GF gradients. To access injectable biomaterials that can mimic such natural electrostatic interactions between soluble signals and macromolecules and mechanically tunable environments, the backbone of agarose, a thermogelling marine–algae-derived polysaccharide, was modified with sulfate, phosphate, and carboxylic moieties and the interaction and release of FGF-2 from these functionalized hydrogels was assessed by ELISA in vitro and CAM assay in ovo. Our findings show that FGF-2 remains active after release, and FGF-2 release profiles can be influenced by sulfated and phosphorylated agarose, and in turn, promote varied blood vessel formation kinetics. These modified agaroses offer a simple approach to mimicking electrostatic interactions experienced by GFs in the extracellular environment and provide a platform to probe the role of these interactions in the modulation of growth factor activity and may find utility as an injectable gel for promoting angiogenesis and as bioinks in 3D bioprinting. Full article
(This article belongs to the Special Issue Biomimetic Drug Delivery Systems 2024)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Chemical structure of the repeat unit of native agarose (NA), carboxylated agarose (CA), sulfated agarose (SA), and phosphorylated agarose (PA). (<b>B</b>) Comparison of the FTIR absorbance spectra of NA and SA. The black arrow points to the vibration of S-O (ν<sub>S = O</sub>) in SA. (<b>C</b>) Comparison of the FTIR absorbance spectra of native agarose (NA) and phosphorylated agarose (PA). The black arrow points to vibrations of POC (ν<sub>POC</sub>) and PO (ν<sub>PO</sub>) in PA. The IR bands at 1158 and 1071 cm<sup>−1</sup> correspond to –C–O–C– and glycosidic linkage [<a href="#B45-biomimetics-10-00012" class="html-bibr">45</a>], while the maxima around 1650 cm<sup>−1</sup> is attributed to polymer-bound water [<a href="#B46-biomimetics-10-00012" class="html-bibr">46</a>].</p>
Full article ">Figure 2
<p>Cumulative H-bond number during the molecular dynamic simulation. Graphs (<b>A</b>–<b>D</b>) show a comparison between H-bonds calculated between two polysaccharide strands and the number of H-bonds between the polysaccharide strands and surrounding water molecules. Graphs (<b>E</b>,<b>F</b>) show a comparison between the interaction potential between strands of the modified agaroses in comparison to native agarose, and how these inter-strand interactions impact H-bonding between the strand and water. Simulations were carried out using the TIP3P water model.</p>
Full article ">Figure 3
<p>(<b>A</b>) Circular dichroism (CD) of 0.1% <span class="html-italic">w</span>/<span class="html-italic">v</span> solution of native agarose (NA), carboxylated agarose (CA), phosphorylated agarose (PA), and sulfonated agarose (SA). (<b>B</b>) Shear modulus (G′) of 2% <span class="html-italic">w</span>/<span class="html-italic">v</span> hydrogels of native agarose (NA), carboxylated agarose (CA), phosphorylated agarose (PA), and sulfated agarose (SA). (<b>C</b>) Scanning electron micrographs of freeze-dried gels prepared from 2% <span class="html-italic">w</span>/<span class="html-italic">v</span> solutions.</p>
Full article ">Figure 4
<p>Mechanism of gelation of the NA (<b>left</b>) depicting the formation of helical bundles due to strong inter-strand H-bond in comparison to SA and PA (<b>right</b>) where the disruption of the helical structures lead to diminished polymer strand interactions and promote H-bonding with water molecules (red) resulting in weak gels.</p>
Full article ">Figure 5
<p>(<b>A</b>) FGF-2 release profile as measured by ELISA from the various modified agarose hydrogels (SA: sulfated agarose; PA: phosphate agarose, CA: carboxylated agarose) showing sustained release for a duration exceeding 1-week with appreciable differences in temporal and cumulative release in comparison to native agarose (NA) (** (<span class="html-italic">p</span> ≤ 0.01)). Data points represent average values with standard deviation (SD). SD bars are only visible in those that exceed size of the size of the symbol. One absorbance unit is 626 pg. All values beyond 10-h were found to be statistically significant with <span class="html-italic">p</span> values of ≤ 0.01 or ≤ 0.001 (see <a href="#biomimetics-10-00012-f0A5" class="html-fig">Figure A5</a> for <span class="html-italic">p</span>-values between various groups and time points) (<b>B</b>) Release profiles during the initial 10-h clearly showing the ability of these hydrogels to sequester FGF-2 to various degree. Release from PA beyond 2 h was found to be statistically significant, from all other conditions, and release from SA was statistically significantly different from CA and NA. (see <a href="#biomimetics-10-00012-f0A5" class="html-fig">Figure A5</a> for <span class="html-italic">p</span>-values between various groups and time points) (<b>C</b>) Release profile beyond 10 h showing linear behavior with similar slop from all hydrogels, suggesting that steady state release of FGF-2 is not impacted by charges characteristic of the hydrogels, implying that dissociation of the FGF-2 from polymer network is the limiting step. (<b>D</b>) The postulated release mechanism of FGF-2 from modified agarose hydrogels. In NA hydrogels (top cartoon) the dense polymer network due to strong interactions between agarose chain limits diffusion, while in the highly charged SA and PA hydrogels (bottom carton) electrostatic sequestration and expulsion dominate.</p>
Full article ">Figure 6
<p>(<b>A</b>) Photographs of vasculature around the different hydrogel on the chorioallantoic membrane (CAM) of a chick egg. (<b>B</b>) Quantification of blood vessels formed around the different hydrogels (native agarose (NA), carboxylated agarose (CA), phosphorylated agarose (PA), and sulfonated agarose (SA)) loaded with FGF-2 and positioned on the CAM over a gelatin mesh. (<b>C</b>–<b>E</b>) Temporal changes in blood vessel numbers showing a higher propensity around SA hydrogels loaded with FGF-2 in comparison to CA and NA, and a similar propensity in comparison to PA hydrogels despite a higher release of FGF-2 from PA hydrogels, suggesting a possible role for sulfonate groups in SA hydrogel in stabilizing FGF-2.</p>
Full article ">Figure A1
<p>FTR spectra of native agarose (NA) and carboxylated agarose (CA). The green arrow indicates the absorption bands at 1158 and 1071 cm<sup>−1</sup> corresponding to –C–O–C– and glycosidic linkage, the peak at 1650 cm<sup>−1</sup> denoted by the black arrow denotes polymer-bound water, and the shoulder at 1750 cm<sup>−1</sup> denoted by the red arrow corresponds to the carbonyl stretching in the carboxylic acid moieties in CA.</p>
Full article ">Figure A2
<p>Visualization of the molecular dynamic simulation at the beginning of the experiment (t = 0 ns) compared to the conformation of the polysaccharide strands at the end of the experiment (t = 15 ns).</p>
Full article ">Figure A3
<p>Zeta potential of the 0.1% <span class="html-italic">w</span>/<span class="html-italic">v</span> solution of native agarose (NA), carboxylated agarose (CA), phosphorylated agarose (PA), and sulfonated agarose (SA).</p>
Full article ">Figure A4
<p>Paired comparison of the FGF-2 release profile over the first 10-h from SA (sulfated agarose) with that from PA (phosphate agarose) (<b>A</b>), CA (carboxylated agarose) (<b>B</b>), and NA (native agarose) (<b>C</b>) hydrogels as measured by ELISA. The plot shows cumulative absorbance as a function of time and the data points represent average values with standard deviation. (* <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure A5
<p><span class="html-italic">p</span>-values for the cumulative release of FGF-2 at various time points between the various hydrogel groups. Significance was assessed using a student’s <span class="html-italic">t</span>-test (paired, one-tailed). A <span class="html-italic">p</span>-value of ≤0.05 was considered statistically significant (* (<span class="html-italic">p</span> ≤ 0.05), ** (<span class="html-italic">p</span> ≤ 0.01), *** (<span class="html-italic">p</span> ≤ 0.001) and **** (<span class="html-italic">p</span> ≤ 0.0001). Gray boxes represent comparisons that were not statistically significant.</p>
Full article ">
10 pages, 3856 KiB  
Case Report
Novel LYST Variants Lead to Aberrant Splicing in a Patient with Chediak–Higashi Syndrome
by Maxim Aleksenko, Elena Vlasova, Amina Kieva, Ruslan Abasov, Yulia Rodina, Michael Maschan, Anna Shcherbina and Elena Raykina
Genes 2025, 16(1), 18; https://doi.org/10.3390/genes16010018 - 26 Dec 2024
Viewed by 375
Abstract
Background: The advent of next-generation sequencing (NGS) has revolutionized the analysis of genetic data, enabling rapid identification of pathogenic variants in patients with inborn errors of immunity (IEI). Sometimes, the use of NGS-based technologies is associated with challenges in the evaluation of the [...] Read more.
Background: The advent of next-generation sequencing (NGS) has revolutionized the analysis of genetic data, enabling rapid identification of pathogenic variants in patients with inborn errors of immunity (IEI). Sometimes, the use of NGS-based technologies is associated with challenges in the evaluation of the clinical significance of novel genetic variants. Methods: In silico prediction tools, such as SpliceAI neural network, are often used as a first-tier approach for the primary examination of genetic variants of uncertain clinical significance. Such tools allow us to parse through genetic data and emphasize potential splice-altering variants. Further variant assessment requires precise RNA assessment by agarose gel electrophoresis and/or cDNA Sanger sequencing. Results: We found two novel heterozygous variants in the coding region of the LYST gene (c.10104G>T, c.10894A>G) in an individual with a typical clinical presentation of Chediak–Higashi syndrome (CHS). The SpliceAI neural network predicted both variants as probably splice-altering. cDNA assessment by agarose gel electrophoresis revealed the presence of abnormally shortened splicing products in each variant’s case, and cDNA Sanger sequencing demonstrated that c.10104G>T and c.10894A>G substitutions resulted in a shortening of the 44 and 49 exons by 41 and 47 bp, respectively. Both mutations probably lead to a frameshift and the formation of a premature termination codon. This, in turn, may disrupt the structure and/or function of the LYST protein. Conclusions: We identified two novel variants in the LYST gene, predicted to be deleterious by the SpliceAI neural network. Agarose gel cDNA electrophoresis and cDNA Sanger sequencing allowed us to verify inappropriate splicing patterns and establish these variants as disease-causing. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>Patient’s visual appearance.</p>
Full article ">Figure 2
<p>Pathognomonic giant inclusions in bone marrow cells. (<b>a</b>) Neutrophil. (<b>b</b>) Promonocyte. (<b>c</b>) Lymphocyte. (<b>d</b>) Eosinophil. Stained with hematoxylin and eosin.</p>
Full article ">Figure 3
<p>Pedigree of the whole family. The black arrow indicates the proband.</p>
Full article ">Figure 4
<p>Pedigree information for family with <span class="html-italic">LYST</span> variants: (<b>a</b>,<b>b</b>) father, (<b>c</b>,<b>d</b>) mother, (<b>e</b>,<b>f</b>) sister, (<b>g</b>,<b>h</b>) patient. Arrows indicate nucleotide substitutions.</p>
Full article ">Figure 5
<p>Agarose gel cDNA electrophoresis revealed the presence of an abnormally shortened cDNA fragment in each variant’s case. HC—healthy control. Ethidium bromide staining.</p>
Full article ">Figure 6
<p>Parents’ cDNA Sanger sequencing. (<b>a</b>) Mother’s cDNA Sanger sequencing: heterozygous c.10894A&gt;G substitution in 49 exon leads to inappropriate splicing of RNA and “loss” of 47 b.p. Normal allele sequence (part of 49 exon) is depicted on a top row, and mutant allele (truncated 49 exon, followed by 50 exon) is depicted on the bottom row. (<b>b</b>) Fathers’ cDNA Sanger sequencing: heterozygous c.10104G&gt;T substitution in 44 exon results in aberrant RNA splicing and “loss” of 41 b.p. Normal allele sequence (part of 44 exon) is depicted on a top row, mutant allele (truncated 44 exon, followed by 45 exon) is depicted on the bottom row.</p>
Full article ">Figure 7
<p>Schematic diagram illustrating abnormal splicing signature: (<b>a</b>) due to c.10894A&gt;G substitution (<b>b</b>) due to c.10104G&gt;T substitution. Constitutive splicing is depicted by green lines; altered splicing is depicted by red lines.</p>
Full article ">
16 pages, 1498 KiB  
Article
The Use of Gel Electrophoresis to Separate Multiplex Polymerase Chain Reaction Amplicons Allows for the Easy Identification and Assessment of the Spread of Toxigenic Clostridioides difficile Strains
by Tomasz Bogiel, Patrycja Kwiecińska, Robert Górniak, Piotr Kanarek and Agnieszka Mikucka
Gels 2024, 10(12), 818; https://doi.org/10.3390/gels10120818 - 12 Dec 2024
Viewed by 503
Abstract
Clostridioides difficile is a common etiological factor of hospital infections, which, in extreme cases, can lead to the death of patients. Most strains belonging to this bacterium species synthesize very dangerous toxins: toxin A (TcdA) and B (TcdB) and binary toxin (CDT). The [...] Read more.
Clostridioides difficile is a common etiological factor of hospital infections, which, in extreme cases, can lead to the death of patients. Most strains belonging to this bacterium species synthesize very dangerous toxins: toxin A (TcdA) and B (TcdB) and binary toxin (CDT). The aim of this study was to assess the suitability of agarose gel electrophoresis separation of multiplex PCR amplicons to investigate the toxinogenic potential of C. difficile strains. Additionally, the frequency of C. difficile toxin genes and the genotypes of toxin-producing strains were determined. Ninety-nine C. difficile strains were used in the detection of the presence of genes encoding all of these toxins using the multiplex PCR method. In 85 (85.9%) strains, the presence of tcdA genes encoding enterotoxin A was detected. In turn, in 66 (66.7%) isolates, the gene encoding toxin B (tcdB) was present. The lowest number of strains tested was positive for genes encoding a binary toxin. Only 31 (31.3%) strains possessed the cdtB gene and 22 (22.2%) contained both genes for the binary toxin subunits (the cdtB and cdtA genes). A relatively large number of the strains tested had genes encoding toxins, whose presence may result in a severe course of disease. Therefore, the accurate diagnosis of patients, including the detection of all known C. difficile toxin genes, is very important. The multiplex PCR method allows for the quick and accurate determination of whether the tested strains of this bacterium contain toxin genes. Agarose gel electrophoresis is a useful tool for visualizing amplification products, allowing one to confirm the presence of specific C. difficile toxin genes as well as investigate their dissemination for epidemiological purposes. Full article
(This article belongs to the Special Issue Gels in Separation Science)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The diversity and number of the detected toxinogenotypes among <span class="html-italic">Clostridioides difficile</span> strains (<span class="html-italic">n</span> = 99), where <span class="html-italic">cdtA</span>+—presence of the gene encoding the A subunit of the binary toxin, <span class="html-italic">cdtA</span>−—no gene present, <span class="html-italic">cdtB</span>+—presence of the gene encoding the B subunit of the binary toxin, <span class="html-italic">cdtB</span>−—no gene present, <span class="html-italic">gluD</span>+—presence of the gene encoding glutamate dehydrogenase, <span class="html-italic">gluD</span>−—no gene present, <span class="html-italic">tcdA</span>+—presence of the gene encoding toxin A, <span class="html-italic">tcdA</span>−—no gene present, <span class="html-italic">tcdB</span>+—presence of the gene encoding toxin B, <span class="html-italic">tcdB</span>−—no gene present.</p>
Full article ">Figure 2
<p>Example of agarose gel electrophoretic separation of the amplification products using the multiplex PCR technique for the <span class="html-italic">tcdA</span> (629 bp), <span class="html-italic">tcdB</span> (410 bp), <span class="html-italic">cdtB</span> (262 bp), <span class="html-italic">cdtA</span> (221 bp), and <span class="html-italic">gluD</span> (158 bp) genes, where <span class="html-italic">cdtA</span>—binary toxin subunit A gene; <span class="html-italic">cdtB</span>—binary toxin subunit B gene; <span class="html-italic">gluD</span>—glutamate dehydrogenase gene; <span class="html-italic">tcdA</span>—toxin A gene; <span class="html-italic">tcdB</span>—toxin B gene; M—DNA size marker 100–1000 base pairs; the lines labelled 40–46, 117—numbers of the tested strains; 42, 46, and K (+)—positive controls of the amplification reaction for all the investigated genes, and the remaining samples with the <span class="html-italic">gluD</span>, <span class="html-italic">tcdA,</span> and <span class="html-italic">tcdB</span> genes only; K (−)—negative control of the amplification reaction.</p>
Full article ">
29 pages, 5745 KiB  
Article
The Effect of a Photoactivated Ruthenium Nitrocomplex [RuCl(NO2)(dppb)(4,4-2 Mebipy)] on the Viability of Eukaryotic and Prokaryotic Cells, Including Bacterial Biofilms
by Alexandre L. Andrade, Aryane A. Pinheiro, Ellen A. Malveira, Isa M. F. Azevedo, Ana C. S. Gondim, Alzir A. Batista, João H. A. Neto, Eduardo E. Castellano, Alda K. M. Holanda, Mayron A. Vasconcelos and Edson H. Teixeira
Processes 2024, 12(12), 2841; https://doi.org/10.3390/pr12122841 - 11 Dec 2024
Viewed by 493
Abstract
Antimicrobial resistance (AMR) is a critical global public health problem. Many bacterial pathogens use biofilm formation as their main pathogenicity mechanism, a practical tactic for surviving in natural settings and colonized host tissues. Research using ruthenium(II) complexes has demonstrated antibacterial action linked to [...] Read more.
Antimicrobial resistance (AMR) is a critical global public health problem. Many bacterial pathogens use biofilm formation as their main pathogenicity mechanism, a practical tactic for surviving in natural settings and colonized host tissues. Research using ruthenium(II) complexes has demonstrated antibacterial action linked to photodynamic therapy, an alternate method of microbial control. Thus, in this work, the photosensitive nitro complex [RuCl(NO2)(dppb)(4,4-Mebipy)] (I) was prepared and the X-ray structure was determined. Then, we investigated the antibacterial and antibiofilm activities, antibiotic-associated effects, and cytotoxicity. The results showed that complex I exhibited promising antimicrobial activity with MIC values ranging from 4 to 256 µg/mL and MBC from 4 to 32 µg/mL. The antimicrobial activity of this nitro complex was significantly enhanced with blue light irradiation, as confirmed by agarose gel electrophoresis of the pBR322 DNA, which must be related to the DNA cleavage promoted by the photorelease of NO. A synergistic effect against Staphylococcus aureus and Staphylococcus epidermidis strains was observed when combined with ampicillin, which exhibited FICI values from 0.186 to 0.311. Interestingly, complex I associated with tetracycline showed a synergistic effect only on Escherichia coli. Regarding biofilms, the irradiated complex I showed antibacterial activity against biofilm formation and mature biofilms. Furthermore, SEM and confocal analyses revealed changes in cell morphology and damage to the wall and plasma membrane. Complex I presented a percentage of hemolysis between 2 and 4%, and no cytotoxic effect was observed against murine dermal fibroblasts. In conclusion, the photoactivated ruthenium(II) complex showed antibacterial and antibiofilm activity against relevant bacteria. Full article
(This article belongs to the Special Issue Microbial Biofilms: Latest Advances and Prospects)
Show Figures

Figure 1

Figure 1
<p>Structure of the nitro complex [RuCl(NO<sub>2</sub>)(dppb)(4,4-Mebipy)], designated complex I.</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) LED light system used for uniform irradiation of samples in a 96-well plate. The system features a dedicated lamp for each well (λmax = 463 nm; power = 8 mW/cm<sup>2</sup>).</p>
Full article ">Figure 3
<p>Molecular geometries for complex I showing the two isomers generated by the disordered NO<sub>2</sub><sup>−</sup> and Cl<sup>−</sup> ligands (ellipsoids at 30% of probability). For the sake of clarity, two methanol molecules as solvate were omitted.</p>
Full article ">Figure 4
<p>Differential pulse voltammogram of complex I in NaTFA solution, pH 7.0; µ = 1 mol/L at a glassy carbon electrode, utilizing a platinum wire as the auxiliary electrode and Ag/AgCl as the reference electrode. The voltammogram was obtained following light irradiation at 463 nm. Solid line: initial substance in the absence of light; dashed line: subsequent to irradiation. The potential scan commenced at 0.3 V with an anodic sweep.</p>
Full article ">Figure 5
<p>UV/Vis/NIR spectroelectrochemical response of complex I during reduction at a negative potential of −1.0 V for 210 min.</p>
Full article ">Figure 6
<p>DNA cleavage assay. Lane 2 shows only pBR322 DNA. Plasmid DNA with complex I at concentrations of 100, 50, and 30 µM was incubated in the dark or exposed to blue light for 1 h. The [Ru(bpy)<sub>3</sub>]<sup>2+</sup> complex served as a positive control for TBP.</p>
Full article ">Figure 7
<p>Effect of Complex I on the biofilm formation (24 h) of <span class="html-italic">S. aureus</span>, <span class="html-italic">S. epidermidis</span>, <span class="html-italic">E. coli</span>, and <span class="html-italic">P. aeruginosa</span>. Quantification of biomass (<b>A</b>–<b>F</b>) by crystal violet (CV) staining assay. White columns denote biofilm subjected to different concentrations of Complex I, while black columns indicate untreated biofilm (control, medium containing 4% DMSO). **** <span class="html-italic">p</span> &lt; 0.0001 represent statistical difference in relation to the control.</p>
Full article ">Figure 8
<p>Effect of Complex I on the number of viable cells present in biofilm formation (24 h) of <span class="html-italic">S. aureus</span>, <span class="html-italic">S. epidermidis</span>, <span class="html-italic">E. coli</span>, and <span class="html-italic">P. aeruginosa</span>. Counting the number of viable cells (<b>A</b>–<b>F</b>). White columns denote biofilm subjected to different concentrations of Complex I, while black columns indicate untreated biofilm (control, medium containing 4% DMSO). **** <span class="html-italic">p</span> &lt; 0.0001 represent statistical difference in relation to the control.</p>
Full article ">Figure 9
<p>Effect of Complex I on the biomass of mature biofilms (48 h) of <span class="html-italic">S. aureus</span>, <span class="html-italic">S. epidermidis</span>, <span class="html-italic">E. coli</span>, and <span class="html-italic">P. aeruginosa</span>. Quantification of biomass (<b>A</b>–<b>F</b>) through crystal violet (CV) staining assay. White columns denote biofilm subjected to different concentrations of Complex I, while black columns indicate untreated biofilm (control, medium containing 4% DMSO). * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001 represent statistical difference in relation to the control.</p>
Full article ">Figure 10
<p>Effect of Complex I on the number of viable cells present in mature biofilms (48 h) of <span class="html-italic">S. aureus</span>, <span class="html-italic">S. epidermidis</span>, <span class="html-italic">E. coli</span>, and <span class="html-italic">P. aeruginosa</span>. Counting the number of viable cells (<b>A</b>–<b>F</b>). White columns denote biofilm subjected to different concentrations of Complex I, while black columns indicate untreated biofilm (control, medium containing 4% DMSO). * <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; 0.0001 represent statistical difference in relation to the control.</p>
Full article ">Figure 11
<p>Electron micrographs obtained by SEM illustrating the impact of Complex I on biofilm development of <span class="html-italic">S. aureus</span> ATCC 700698 (<b>A</b>–<b>D</b>) and <span class="html-italic">E. coli</span> ATCC 11303 (<b>E</b>–<b>H</b>) following 24 h of treatment. Electron micrographs show untreated biofilms of <span class="html-italic">S. aureus</span> (<b>A</b>,<b>B</b>) and <span class="html-italic">E. coli</span> (<b>E</b>,<b>F</b>) at magnifications of 20,000× and 65,000×, respectively. Electron micrographs show biofilms of <span class="html-italic">S. aureus</span> (<b>C</b>,<b>D</b>) and <span class="html-italic">E. coli</span> (<b>G</b>,<b>H</b>) treated with Complex I at a concentration of 32 μg/mL and 256 μg/mL, respectively, at magnifications of 20,000× and 65,000×.</p>
Full article ">Figure 12
<p>Micrographs of the evaluation of the effect of Complex I on biofilm formation (24 h) and mature biofilms (48 h) of <span class="html-italic">S. aureus</span> ATCC 700698. (<b>A</b>) Untreated <span class="html-italic">S. aureus</span> ATCC 700698 (MRSA) biofilms. (<b>B</b>) <span class="html-italic">S. aureus</span> ATCC 700698 (MRSA) biofilms treated with Complex I at a concentration of 32 μg/mL. (<b>C</b>) Untreated <span class="html-italic">S. aureus</span> ATCC 700698 (MRSA) mature biofilms. (<b>D</b>) <span class="html-italic">S. aureus</span> ATCC 700698 (MRSA) mature biofilms treated with Complex I at a concentration of 64 μg/mL. Magnitude: 600× (<b>A</b>–<b>D</b>). Lasers: 488 nm for SYTO9 detection and 561 nm for propidium iodide detection.</p>
Full article ">Figure 13
<p>Hemolytic activity of Complex I on human erythrocytes. **** <span class="html-italic">p</span> &lt; 0.0001 represents statistical differences concerning the total hemolysis control (SDS 1%).</p>
Full article ">Figure 14
<p>Cytotoxicity of nitro complex I against murine fibroblast L929 cell line. The cell viability was determined by MTT assay. After (<b>A</b>) 24 h and (<b>B</b>) 48 h of treatment. Error bars indicate standard error of the mean (SEM). *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 compared to control (medium with 1% DMSO).</p>
Full article ">
11 pages, 1342 KiB  
Article
Acinetobacter baumannii Co-Resistant to Extended Spectrum Beta-Lactamases and Carbapenemases in Six Peruvian Hospital Centers
by Mabel R. Challapa-Mamani, José Yareta, Alexander Fajardo-Loyola, Percy Asmat Marrufo, Carlos Peralta Siesquen, Jimena Pino-Dueñas, Henry Meza-Fernández, Jhony A. De La Cruz-Vargas and Pool Marcos-Carbajal
Microbiol. Res. 2024, 15(4), 2650-2660; https://doi.org/10.3390/microbiolres15040175 (registering DOI) - 11 Dec 2024
Viewed by 584
Abstract
Objectives: The aim of this study was to describe the phenotypic and molecular characteristics of Acinetobacter baumannii isolates carrying resistance genes to beta-lactams and carbapenems in six Peruvian public hospital centers. Materials and methods: The susceptibility of bacterial isolates was determined using the [...] Read more.
Objectives: The aim of this study was to describe the phenotypic and molecular characteristics of Acinetobacter baumannii isolates carrying resistance genes to beta-lactams and carbapenems in six Peruvian public hospital centers. Materials and methods: The susceptibility of bacterial isolates was determined using the automated MicroScan system, with interpretation according to the M100 S30 CLSI 2020. Resistance genes were identified by conventional polymerase chain reaction (PCR), and PCR products were visualized by 1% agarose gel electrophoresis. Results: Nine strains (TRU1, PM1, PM2, CUS1, CUS2, CUS3, CAL1, CAL2 and CAL3) out of a total of 21 strains in the study were reactivated, showing resistance of 77.8% to imipenem, ciprofloxacin and cefepime, followed by 66.7% resistance to meropenem and ceftazidime, indicating marked multidrug resistance. In addition, the detection of the group A beta-lactamase genes blaCTX-M and blaTEM was confirmed, showing co-resistance in strains CUS1, CUS2 and CUS3, despite their unusual presence in this pathogen, also determined by the presence of the group D carbapenemase blaOXA in strain CUS3, the only strain to show co-resistance of the three groups. Conclusion: The prevalence of Acinetobacter baumannii resistant to extended-spectrum beta-lactamases and carbapenemases in Peruvian public centers represents a critical challenge for the treatment of infections. Rigorous surveillance, infection control strategies, and the development of alternative therapies are urgently needed to address this growing bacterial resistance. Full article
Show Figures

Figure 1

Figure 1
<p>Frequency of antimicrobial resistance in <span class="html-italic">A. baumannii</span> strains producing resistance genes. CAZ: ceftazidime; FEP: cefepime; IPM: imipenem; MEM: meropenem; AMK: amikacin; GEN: gentamicin; TOB: tobramycin; CIP: ciprofloxacin; SXT: sulfamethoxazole/trimethoprim.</p>
Full article ">Figure 2
<p>Number of <span class="html-italic">A. baumannii</span> samples with resistance genes detected and electrophoretic run photo for the ctx gene. CTX-M: cefotaxime; TEM: temoniera; OXA: oxacillinase; bla: beta-lactamase genes.</p>
Full article ">
13 pages, 2254 KiB  
Article
Activation Energy of SDS–Protein Complexes in Capillary Electrophoresis with Tetrahydroxyborate Cross-Linked Agarose Gels
by Dániel Sárközy and András Guttman
Gels 2024, 10(12), 805; https://doi.org/10.3390/gels10120805 - 7 Dec 2024
Viewed by 582
Abstract
Hydrogels like agarose have long been used as sieving media for the electrophoresis-based analysis of biopolymers. During gelation, the individual agarose strands tend to form hydrogen-bond mediated double-helical structures, allowing thermal reversibility and adjustable pore sizes for molecular sieving applications. The addition of [...] Read more.
Hydrogels like agarose have long been used as sieving media for the electrophoresis-based analysis of biopolymers. During gelation, the individual agarose strands tend to form hydrogen-bond mediated double-helical structures, allowing thermal reversibility and adjustable pore sizes for molecular sieving applications. The addition of tetrahydroxyborate to the agarose matrix results in transitional chemical cross-linking, offering an additional pore size adjusting option. Separation of SDS-proteins during gel electrophoresis is an activated process defined by the interplay between viscosity, gelation/cross-link formation/distortion, and sample conformation. In this paper, the subunits of a therapeutic monoclonal antibody were separated by capillary SDS agarose gel electrophoresis at different temperatures. The viscosity of the separation matrix was also measured at all temperatures. In both instances, Arrhenius plots were used to obtain the activation energy values. It was counterintuitively found that larger SDS–protein complexes required lower activation energies while their low-molecular-weight counterparts needed higher activation energy for their electromigration through the sieving matrix. As a first approximation, we considered this phenomenon the result of the electric force-driven distortion of the millisecond range lifetime reticulations by the larger and consequently more heavily charged electromigrating molecules. In the meantime, the sieving properties of the gel were still maintained, i.e., they allowed for the size-based separation of the sample components, proving the existence of the reticulations. Information about the activation energy sheds light on the possible deformation of the sieving matrix and the solute molecules. In addition, the activation energy requirement study helped in optimizing the separation temperature, e.g., with our sample mixture, the highest resolution was obtained for the high-molecular-weight fragments, i.e., between the non-glycosylated heavy chain and heavy-chain subunits at 25 °C (lower Ea requirement), while 55 °C was optimal for the lower-molecular-weight light chain and non-glycosylated heavy chain pair (lower Ea requirement). Future research directions and possible applications are also proposed. Full article
(This article belongs to the Section Gel Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic representation of the di–diol (2:1) linkage formation during agarose gel cross-linking with tetrahydroxyborate.</p>
Full article ">Figure 2
<p>Reproducibility of SDS capillary agarose gel electrophoresis separation of the subunits of omalizumab using tetrahydroxyborate cross-linker. The overlay of ten consecutive runs demonstrates reproducible peak alignment, with all peaks showing consistent migration times and peak shapes. Peaks: (1) 10 kDa internal standard, (2) light-chain subunit, (3) non-glycosylated heavy-chain subunit, and (4) heavy-chain subunit of omalizumab. Conditions: sieving matrix, 0.8% (<span class="html-italic">w</span>/<span class="html-italic">v</span>) agarose in TBEG background electrolyte (pH 7.0); capillary, 20 cm effective length 50 µm i.d. bare fused silica (total length: 30 cm); detection, 214 nm ultraviolet light absorption; applied voltage, −15 kV (anode at the detection side); separation temperature, 25 °C ± 0.1 °C; electrokinetic injection, −5 kV for 10 s.</p>
Full article ">Figure 3
<p>The effect of temperature on the SDS capillary agarose gel electrophoresis separation of the subunits of the therapeutic monoclonal antibody omalizumab, using tetrahydroxyborate cross-linker. Traces: (<b>a</b>) 25 °C, (<b>b</b>) 35 °C, (<b>c</b>) 45 °C and (<b>d</b>) 55 °C. Peaks: (1) 10 kDa internal standard, (2) LC, (3) ngHC, and (4) HC fragments. Separation conditions are the same as in <a href="#gels-10-00805-f002" class="html-fig">Figure 2</a>, except for the temperatures of the analyses.</p>
Full article ">Figure 4
<p>Arrhenius plots of logarithmic mobility vs. reciprocal absolute temperature of the SDS–protein complexes in the sample mixture. The symbols representing the sample molecules are defined in the inset. The error bars show the standard deviation of 3 parallel experiments.</p>
Full article ">Figure 5
<p>Relationship between the activation energies (derived from the slopes of <a href="#gels-10-00805-f004" class="html-fig">Figure 4</a>) and the molecular weights of the separated SDS–protein complexes. The error bars represent the standard deviation of 3 parallel experiments.</p>
Full article ">Figure 6
<p>Logarithmic viscosity vs. reciprocal absolute temperature (Arrhenius) plot representing the temperature-dependent viscosity changes of the borate cross-linked agarose gels (r<sup>2</sup> = 0.984).</p>
Full article ">
18 pages, 11305 KiB  
Article
Preparation and Evaluation of a Combination of Chelating Agents for the Removal of Inhaled Uranium
by Lintao Li, Runtian Li, Ruohan Guo, Shuang Guo, Xuan Qiao, Xinru Wu, Peng Han, Yunbo Sun, Xiaoxia Zhu, Zhuona Wu, Hui Gan, Zhiyun Meng, Guifang Dou, Ruolan Gu and Shuchen Liu
Molecules 2024, 29(23), 5759; https://doi.org/10.3390/molecules29235759 - 5 Dec 2024
Viewed by 503
Abstract
Inhalation of aerosolized uranium is recognized as a principal mode of exposure, posing significant risks of damage to the lungs, kidneys, and other vital organs. To enhance nuclide elimination from the body, chelating agents are employed; however, single-component chelators often exhibit limited spectral [...] Read more.
Inhalation of aerosolized uranium is recognized as a principal mode of exposure, posing significant risks of damage to the lungs, kidneys, and other vital organs. To enhance nuclide elimination from the body, chelating agents are employed; however, single-component chelators often exhibit limited spectral activity and low effectiveness, resulting in toxicologically relevant concentrations. We have developed a composite chelating agent composed of 3,4,3-Li(1,2-HOPO), DFP, and HEDP in optimized ratios, demonstrating marked improvements in eliminating inhaled uranium. The selection of these components was initially guided by an agarose gel dynamics method, focusing on uranium binding and removal efficacy. Optimization of the formula was conducted through response surface methodology in a cellular model. The compound’s ability to enhance survival rates in mice subjected to acute uranium inhalation was confirmed, showing a dose-dependent improvement in survival in severely affected mice. Comparative assessments indicated that this multifaceted chelating agent substantially surpasses the uranium tissue clearance achieved by individual chelating agents. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Some of the chelating ligands’ structures are shown in the figure.</p>
Full article ">Figure 2
<p>Uranyl nitrate was included in the static retention phase prepared in NS. After 5 min (<b>B</b>), 15 min (<b>C</b>), 30 min (<b>D</b>), 1 h (<b>E</b>), and 2 h (<b>F</b>) incubation containing different chelating agents, the nuclide transfer rate was measured using ICP-MS (Model 7900, Agilent, Santa Clara, CA, USA). (<b>A</b>) show the trends in nuclide transfer rates of different types of chelating agents over time. Results are expressed as the content in the mobile phase/total content in the gel × 100. * <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 is compared to the most significant group exposed to blank. Each experimental condition was performed in triplicate. Bars represent mean ± sd.</p>
Full article ">Figure 3
<p>U<sub>3</sub>O<sub>8</sub> was included in the static retention phase prepared in NS. After 5 min (<b>B</b>), 15 min (<b>C</b>), 30 min (<b>D</b>), 1 h (<b>E</b>), and 2 h (<b>F</b>) incubation containing different chelating agents, the nuclide transfer rate was measured using ICP-MS. (<b>A</b>) show the trends in nuclide transfer rates of different types of chelating agents over time. Results are expressed as the content in the mobile phase/total content in the gel × 100. * <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 is compared to the most significant group exposed to blank. Each experimental condition was performed in triplicate. Bars represent mean ± sd.</p>
Full article ">Figure 4
<p>Cytotoxicity assay results. The upper limit of the dosing concentration (Uranyl nitrate (<b>A</b>), HOPO (<b>B</b>), DFP (<b>C</b>), HEDP (<b>D</b>)) was set at a cell viability rate of greater than 75%. Each concentration was repeated six times.</p>
Full article ">Figure 5
<p>The removal efficiency of uranium of different chelating agents at different concentrations. HOPO (<b>A</b>), DFP (<b>B</b>), and HEDP (<b>C</b>), at different concentrations (µg/mL) on BEAS-2B cells exposed to U(VI). * <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 is compared to the significant group exposed to U(VI). Bars represent mean ± sd.</p>
Full article ">Figure 6
<p>Graphical results in terms of the response surfaces of the format value of Y2. Part (<b>A</b>): Joint graphical 3D analysis as a function of each of the variables involved. Each of the net surfaces represents the theoretical three-dimensional response surface predicted with the second-order polynomial as a function of each one of the involved variables. The statistical design and results are described in <a href="#molecules-29-05759-t002" class="html-table">Table 2</a>. The estimated parametric values are shown in <a href="#molecules-29-05759-t003" class="html-table">Table 3</a>. Part (<b>B</b>): RSM contour charts. From red to blue, the response variable value is from high to low.</p>
Full article ">Figure 7
<p>Mice survival rate experiment (<b>A</b>) and weight change (<b>B</b>) during 14d. Each mice received uranyl nitrate in the lungs and was then either treated promptly or left untreated 10 min after uranium exposure, according to the regimens described above.</p>
Full article ">Figure 8
<p>The depletion of uranium in rats was achieved through intratracheal injection after 10 min exposure of single or a combination of chelating agents, with efficacy observed 24 h after exposure. The figure illustrates the uranium content in the tissue (lung (<b>A</b>), kidney (<b>B</b>), liver (<b>C</b>), bone (<b>D</b>)). * <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 is compared to the MIX group (<b>A</b>–<b>C</b>) or the control group(<b>D</b>) exposed to U(VI). Bars represent mean ± sd.</p>
Full article ">Figure 9
<p>Twenty-four hours following uranyl nitrate poisoning, pathological alterations to lung tissue were observed. Lung and kidney tissue specimens from each group (n = 3) were examined under an optical microscope after HE staining (HE 20×). The experimental groups were identical to those previously described. The significance of the scores (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01) is then compared with the control group. Black arrows, alveolar cavities demonstrated atrophy; green arrow, inflammatory cells infiltrated in the alveolar septum and red arrow, red blood cells observed in alveolar cavities.</p>
Full article ">
8 pages, 3070 KiB  
Communication
A Novel Polymerase Chain Reaction (PCR)-Based Method for the Rapid Identification of Chrysodeixis includens and Rachiplusia nu
by Guilherme A. Gotardi, Natália R. F. Batista, Tamylin Kaori Ishizuka, Luiz H. Marques, Mário H. Dal Pogetto, Amit Sethi, Mark L. Dahmer and Timothy Nowatzki
Insects 2024, 15(12), 969; https://doi.org/10.3390/insects15120969 - 4 Dec 2024
Viewed by 766
Abstract
Chrysodeixis includens and Rachiplusia nu are two species belonging to the Plusiinae subfamily within the Noctuidae family. Due to their morphological similarity, the identification of their larvae is difficult and time-consuming. A rapid and accurate identification of these two species is essential for [...] Read more.
Chrysodeixis includens and Rachiplusia nu are two species belonging to the Plusiinae subfamily within the Noctuidae family. Due to their morphological similarity, the identification of their larvae is difficult and time-consuming. A rapid and accurate identification of these two species is essential for their management as these species exhibit differential susceptibilities to insecticides and crops engineered to express Bacillus thuringiensis (Bt) proteins, and a molecular tool can easily provide this differentiation. Currently, molecular analysis can identify these species through genetic sequencing, an expensive and time-consuming process. In our study, after sequencing part of the mtDNA cytochrome c oxidase I (COI) gene and based on the differences found in the gene of each species, a set of species-specific primers was developed: one reverse primer common to both species and two forward primers, specific to each species, amplifying fragments of 199 base pairs (bp) for C. includens and 299 bp for R. nu. Our results indicate that the primers were specific for these species, enabling the identification of individuals directly through agarose gel. The new methodology proved to be accurate, rapid, and reliable for the correct identification of these two species of loopers. Full article
(This article belongs to the Section Insect Molecular Biology and Genomics)
Show Figures

Figure 1

Figure 1
<p>Alignment of partial mtCOI gene sequences for <span class="html-italic">C. includens</span>, <span class="html-italic">R. nu</span>, <span class="html-italic">Trichoplusia ni</span>, <span class="html-italic">A. gamma</span>, and <span class="html-italic">A. egena</span>. The black dots indicate the nucleotide positions conserved across all the species. The nucleotides in white text over black backgrounds highlight differences between <span class="html-italic">C. includens</span> and <span class="html-italic">R. nu</span>. The sequences underlined in red indicate the position of the primers.</p>
Full article ">Figure 2
<p>Multiple sequence alignment of the mitochondrial cytochrome c oxidase subunit I (mtCOI) gene region for <span class="html-italic">R. nu</span> and <span class="html-italic">C. includens</span>, highlighting nucleotide variability between the species. The reference sequences used were KC354734.1 for <span class="html-italic">R. nu</span> and MT180766.1 for <span class="html-italic">C. includens</span>. Identical nucleotides are displayed in white text over a black background, while mismatched nucleotides are shown in plain text.</p>
Full article ">Figure 3
<p>SYBR Safe-stained 2% agarose gel electrophoresis of mtCOI PCR products. Lanes 1–5 show a 199 bp amplicon confirming <span class="html-italic">C. includens</span>. Lanes 6–10 show a 299 bp amplicon indicating <span class="html-italic">R. nu</span>. Lane M contains a 1 kb DNA ladder for size estimation, and lane N is a negative control.</p>
Full article ">Figure 4
<p>Agarose gel electrophoresis of mtCOI gene amplicons for validating method with field-collected insect samples. M: 1 kb DNA ladder; Lanes 1–6, 9, 10, and 12–19: 299 bp amplicon for <span class="html-italic">R. nu</span>; Lanes 7, 8, and 11: 199 bp amplicon for <span class="html-italic">C. includens</span>.</p>
Full article ">
14 pages, 4762 KiB  
Article
Immunohistochemical Localization and Expression of Aquaporin 3-2 (AQP3C1) in the Spiny Dogfish, Squalus acanthias
by Christopher P. Cutler and Tolulope Ojo
Hydrobiology 2024, 3(4), 378-391; https://doi.org/10.3390/hydrobiology3040024 - 4 Dec 2024
Viewed by 617
Abstract
The previously available coding region for the spiny dogfish (Squalus acanthias) AQP3-2 gene was amplified from cDNAs using PCR. Agarose gel electrophoresis gave a band of the AQP3-2 coding region, as well as multiple smaller splice variant bands. The main AQP3-2 [...] Read more.
The previously available coding region for the spiny dogfish (Squalus acanthias) AQP3-2 gene was amplified from cDNAs using PCR. Agarose gel electrophoresis gave a band of the AQP3-2 coding region, as well as multiple smaller splice variant bands. The main AQP3-2 band and the largest and most fluorescently intense pair of these splice variant bands were cloned and sequenced. Amplifications were performed on a range of tissue cDNAs, but AQP3-2 was only expressed in the kidney and brain. Quantitative PCR amplifications using pre-existing kidney cDNA from an environmental salinity acclimation experiment showed that the abundance of mRNA from both the main AQP3-2 transcript and the largest splice variant (Splice Variant 1) was lower in 120% seawater (SW) acclimated fish, although only the values for Splice Variant 1 were statistically significant. A custom-made affinity-purified rabbit polyclonal AQP3-2 antibody was produced, and this gave four bands of around the correct sizes (which were 27 and 32 kDa) for the complete AQP3-2 and Splice Variant 1 proteins. Two of the bands may have been N-glycosylated forms of these proteins. Other bands were also present on the Western blot. No bands were present when the antibody was pre-blocked by the peptide antigen. In tissue sections of the dogfish kidney, immunohistochemical localization experiments showed that AQP3-2 was expressed in the early distal tubule (EDT) and late distal tubule (LDT) nephron segments. The results suggest that AQP3-2 may be involved in cell volume regulation in the EDT and water and urea absorption in the LDT nephron segment. Full article
Show Figures

Figure 1

Figure 1
<p>Agarose electrophoresis gel of the PCR amplifications of the AQP3-2 gene coding region using various tissue (as shown) cDNAs from the spiny dogfish <span class="html-italic">Squalus acanthias</span>. The expected size of the full-length coding region was 954 bp, based on the pre-existing sequence in the genebank (Accession Number, HAGV01050720; [<a href="#B11-hydrobiology-03-00024" class="html-bibr">11</a>]). The band sizes shown on the right were determined by cloning and sequencing.</p>
Full article ">Figure 2
<p>Alignment of <span class="html-italic">Squalus acanthias</span> amino acid sequences derived from the aquaporin 3-2 (AQP3-2) cDNA sequence, in comparison to that of aquaporin 3 (AQP3; genebank Accession Number MZ508340, also see [<a href="#B10-hydrobiology-03-00024" class="html-bibr">10</a>]). Single underlining indicates the peptide regions used for antibody production. Double underlining indicates the region of amino acids missing in AQP3-2 Splice Variant 1. Bolding indicates the single potential N-glycosylation sites within both AQP3 and AQP3-2. Vertical lining indicates identical amino acids in the two sequences. Double dots indicate chemically similar amino acids in each sequence. Single dots indicate less similar amino acids in each sequence. A dash indicates an amino acid missing in comparison to the other sequence. Both sequences contain the two conserved NPA motifs characteristic of aquaporins.</p>
Full article ">Figure 3
<p>Quantitative PCR (QPCR) measurements of mRNA/cDNA expression in the kidney of spiny dogfish. (<b>A</b>) AQP3-2 and the splice variant; (<b>B</b>) AQP3-2 Splice Variant 1 in fish acclimated to 75% seawater (SW) or 120% SW compared to 100% SW controls. ** <span class="html-italic">p</span> =&lt; 0.01, *** <span class="html-italic">p</span> =&lt; 0.001. Error bars are +/− standard error of the mean.</p>
Full article ">Figure 4
<p>Western blot using the AQP3-2 A1 antibody. A 300 μg protein sample of purified plasma membranes from the kidney was run on a 10% SDS polyacrylamide gel. The blots were incubated with either the rabbit-anti dogfish AQP3-2 primary antibody (Kidney lane) or with the same antibody pre-blocked for &gt;1 h with 50 μg/mL peptide antigen (Kidney-Peptide Blocked lane). M = Precision Plus Protein Kaleidoscope marker (Biorad, Santa Rosa, CA, USA). Antibody detection was performed using a donkey anti-rabbit IgG secondary antibody with conjugated alkaline phosphatase enzyme and NBT/BCIP chromogenic alkaline phosphatase substrate (both Peirce, Waltham, MA, USA).</p>
Full article ">Figure 5
<p>Preliminary Zeiss LSM710 confocal microscope images of the kidney stained with the AQP3-2 A1 (<b>A</b>,<b>C</b>) and A2 (<b>B</b>,<b>D</b>–<b>H</b>) antibodies labeled with an Alexa 488 dye (green). Images (<b>A</b>,<b>B</b>) are sections (20× lens) from the sinus zone of the kidney. Sections (<b>C</b>,<b>D</b>) are serial sections (40× lens) from the bundle zone, showing a collection of five EDT nephron tubules together. Sections (<b>E</b>–<b>H</b>) are images taken using the microscope’s Aryscan enhanced resolution module. (<b>E</b>) (40× oil lens) is probably a section through the EDT tubule at the end of the second bundle zone loop. (<b>F</b>) is a magnified part from section (<b>E</b>) (100× oil lens). (<b>G</b>) shows a cross-section through part of a convoluted bundle zone EDT nephron tubule (40× oil lens). (<b>H</b>) shows a cross-section through a stained nephron tubule in the sinus zone. All tubules show basolateral staining with the AQP3-2 antibodies. Staining with the A1 and A2 antibodies was essentially the same. The sections were counterstained with the nuclear dye Dapi (blue). The scale bars show the actual magnification of each image.</p>
Full article ">Figure 6
<p>Control sections regarding the AQP3-2 antibody. Sections (<b>A</b>,<b>B</b>,<b>E</b>,<b>F</b>); A1 antibody. Sections (<b>C</b>,<b>D</b>,<b>G</b>,<b>H</b>); A2 antibody. Sections (<b>A</b>,<b>C</b>,<b>E</b>) were stained with the AQP3-2 antibodies (Alexa 488 dye; green) and are of the kidney sinus zone, the straight bundle zone, and the convoluted bundle zone, respectively. Sections (<b>B</b>,<b>D</b>,<b>F</b>) are comparable (to <b>A</b>,<b>C</b>,<b>E</b>) serial sections with AQP3-2 antibodies pre-blocked with the peptide antibody 50 μg/mL &gt; 1 h. In sections (<b>A</b>,<b>B</b>), the corresponding nephron tubules are labeled with numbers 1-5. In sections (<b>C</b>–<b>F</b>), the corresponding nephron tubules are labeled with EDT on each of the pairs of sections ((<b>C</b>)/(<b>D</b>) and (<b>E</b>)/(<b>F</b>)). Sections (<b>G</b>) (sinus zone) and (<b>H</b>) (convoluted bundle zone) are sections incubated with the AQP3-2 antibody pre-blocked with the AQP3 antibody peptide antigen (see <a href="#hydrobiology-03-00024-f002" class="html-fig">Figure 2</a> and [<a href="#B9-hydrobiology-03-00024" class="html-bibr">9</a>,<a href="#B10-hydrobiology-03-00024" class="html-bibr">10</a>]). As can be seen, there was no perceptible effect of AQP3 peptide antigen blocking on the AQP3-2 antibody fluorescent signals. The sections were counterstained with the nuclear dye Dapi (blue). The scale bars show the actual magnification of each image.</p>
Full article ">Figure 7
<p>Sections (<b>A</b>,<b>B</b>) (20× lens) are serial sections from the kidney sinus zone and were incubated with the AQP3-2 A1 antibody (Alexa 488; green). (<b>C</b>,<b>D</b>) (40× lens) are from the renal convoluted bundle zone and were incubated with the AQP3-2 A2 antibody (Alexa 488; green). All sections were also labeled with a mouse anti-acetylated tubulin antibody, detected with a highly cross-absorbed anti-mouse secondary antibody labeled with Dylight 550 (orange). Sections (<b>A</b>,<b>C</b>) were also labeled with AQP3 antibody and sections (<b>B</b>,<b>D</b>) were labeled with a AQP4/2 antibody, both of which were directly labeled with a CF633 dye (Mix-n-Stain kit, Biotium, Fremont, CA, USA; Red; see [<a href="#B9-hydrobiology-03-00024" class="html-bibr">9</a>,<a href="#B10-hydrobiology-03-00024" class="html-bibr">10</a>]). Section (<b>E</b>) represents the AQP3-2 antibody staining (green) on its own, as seen in section (<b>A</b>). Section (<b>F</b>) represents the AQP3-2 antibody staining (green) on its own, as seen in section (<b>B</b>). mLDT = middle region of the LDT nephron segment. sLDT = start of the LDT tubule segment. The tubulin antibody, which labels cilia, is used to differentiate between the similar IS-I (with cilia) and LDT (without cilia) tubules. The sections were counterstained with the nuclear dye Dapi (blue). The scale bars show the actual magnification of each image.</p>
Full article ">Figure 8
<p>Section (<b>B</b>) is a panel (20× lens) of the sinus zone of the kidney stained with the AQP3-2 A1 antibody (green), the AQP3 antibody (red), the anti-tubulin antibody (orange) that stains cilia, and the nuclear counter stain Dapi (blue). Section (<b>A</b>) shows the AQP3-2 antibody staining (green) on its own, as seen in section (<b>B</b>). Section (<b>C</b>) shows the AQP3 antibody staining (red) on its own, as seen in section (<b>B</b>). The scale bars show the actual magnification of each image.</p>
Full article ">
11 pages, 1253 KiB  
Article
Development of Functional Molecular Markers for Viviparous Germination Resistance in Rice
by So-Myeong Lee, Youngho Kwon, Sung-Ryul Kim, Ju-Won Kang, Hyeonjin Park, Jin-Kyung Cha, Dong-Soo Park, Jun-Hyun Cho, Woojae Kim, Gyu-Hyeon Eom and Jong-Hee Lee
Agronomy 2024, 14(12), 2896; https://doi.org/10.3390/agronomy14122896 - 4 Dec 2024
Viewed by 433
Abstract
Rice (Oryza sativa) plays a pivotal role in global food security. Understanding the genetics of rice cultivation is crucial, particularly for traits such as viviparous germination, which significantly influences germination and yield. Our research aimed to elucidate the genetic and molecular [...] Read more.
Rice (Oryza sativa) plays a pivotal role in global food security. Understanding the genetics of rice cultivation is crucial, particularly for traits such as viviparous germination, which significantly influences germination and yield. Our research aimed to elucidate the genetic and molecular mechanisms by which the Sdr4 gene influences viviparous germination and to develop novel molecular markers for this gene to enhance breeding strategies against viviparous germination. In all, 683 rice cultivars and 100 F2 plants were used for viviparous germination and genetic analysis using KASP (Kompetitive Allele-Specific PCR) and agarose gel-based markers related to viviparous germination tolerance. We developed and used a polymorphic agarose gel-based marker and a KASP marker targeting the Sdr4 gene. A genetic analysis of field-grown rice cultivars and the F2 population revealed that the two markers on Sdr4 were functional for the genomic selection of SNPs and InDels related to dormancy. The Pearson correlation coefficient (r = 0.74, p-value = 3.31 × 10−8) between the Sdr4-IND KASP marker genotype and viviparous germination rate demonstrated a significant positive correlation, supporting the marker’s utility for selecting rice varieties with diminished viviparous germination. This insight serves as a critical theoretical foundation for breeding strategies for developing early-maturing rice varieties with enhanced resistance to viviparous germination, addressing pivotal challenges in rice cultivation, and ensuring food security. Full article
(This article belongs to the Special Issue Genetics and Breeding of Field Crops in the 21st Century)
Show Figures

Figure 1

Figure 1
<p>The results of the <span class="html-italic">Sdr4-dm</span> marker experiment. (<b>a</b>) An electrophoresis image of the <span class="html-italic">Sdr4-dm</span> InDel marker with a positive control; bands appear only in individuals with the <span class="html-italic">Sdr4-k</span> homozygous allele and heterozygous genotype, as the marker is dominant. (<b>b</b>) Phenotypic differences observed between individuals with the <span class="html-italic">Sdr4-k</span> and <span class="html-italic">Sdr4-n</span> alleles were noted after incubating the seeds at 32 °C and 100% relative humidity for 7 days. (<b>c</b>) An assessment of the relationship between the <span class="html-italic">Sdr4</span> genotype and viviparous germination rates in the Saeilmi × NRT383 F<sub>2</sub> population (circles represent outliers). Double asterisks (**) indicate a significant difference at the 1% significance level in viviparous germination rates between individuals with the <span class="html-italic">Sdr4-k</span> and <span class="html-italic">Sdr4-n</span> alleles.</p>
Full article ">Figure 2
<p>A distribution plot of alleles for the <span class="html-italic">Sdr4</span>-IND KASP marker in the Saeilmi × NRT383 F<sub>2</sub> population. Blue dots represent individuals with the <span class="html-italic">Sdr4-n</span> homozygous allele, green dots indicate heterozygous individuals, and red dots represent individuals with the <span class="html-italic">Sdr4-k</span> homozygous allele. The two black squares on the bottom left represent the NTC (No-Template Control), which ensures there is no contamination or non-specific amplification during the KASP marker assay.</p>
Full article ">
15 pages, 2611 KiB  
Article
Variable Assembly and Procapsid Binding of Bacteriophage P22 Terminase Subunits in Solution
by Julia Elise Cabral, Yanfei Qiu, Albert J. R. Heck and Reginald McNulty
Pathogens 2024, 13(12), 1066; https://doi.org/10.3390/pathogens13121066 - 3 Dec 2024
Viewed by 658
Abstract
Concatemeric viral DNA is packaged into bacteriophage P22 procapsids via a headful packaging mechanism mediated by a molecular machine consisting of small (gp3) and large (gp2) terminase subunits. Although a negative stain reconstruction exists for the terminase holoenzyme, it is not clear how [...] Read more.
Concatemeric viral DNA is packaged into bacteriophage P22 procapsids via a headful packaging mechanism mediated by a molecular machine consisting of small (gp3) and large (gp2) terminase subunits. Although a negative stain reconstruction exists for the terminase holoenzyme, it is not clear how this complex binds the dodecameric portal protein located at a 5-fold mismatch vertex. Herein, we describe new assemblies for the holoenzyme. Both native mass spectrometry and transmission electron microscopy reveal that the P22 terminase complex adopts three main assemblies, which include a nonameric S-terminase bound to two L-terminase 1(gp3)9:2(gp2), two nonameric S-terminase bound to five L-terminase 2(gp3)9:5(gp2), and three nonameric S-terminase bound to seven L-terminase 3(gp3)9:7(gp2). Native agarose gel electrophoresis shows that the terminase complex interacts with procapsids with mild crosslinking. These results herein illustrate the P22 terminase complex can adopt a variety of conformations and assembly states. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic of bacteriophage P22 maturation. (1) Nucleation involves the portal protein (gp1), scaffold protein (gp8), injection proteins (gp7, gp16, and gp20), and coat protein (gp5). (2) A spherical procapsid forms with a dodecameric portal protein located at a symmetry-mismatched 5-fold vertex. (3) The large (gp2) and small (gp3) terminase complex binds to the pac site on P22 DNA. The terminase complex utilizes ATPase activity to package concatemeric DNA inside the procapsid, causing rearrangement of the coat proteins. Completion of headful packaging results in DNA cleavage via the gp2 nuclease domain. (4) Tail adapter proteins gp4 and gp10 bind to the portal vertex. (5) Tailspike (gp9) and needle protein (gp26) form the P22 tail. (6) The mature infectious particle is now capable of binding to the <span class="html-italic">Salmonella</span> membrane. Injection proteins (gp7, gp16, and gp20) form a tube, facilitating the safe delivery of the P22 genome into <span class="html-italic">Salmonella</span>.</p>
Full article ">Figure 2
<p>Co-expression and purification of the gp3:gp2 complex. (<b>A</b>) Domain organization of P22 S- and L-terminase subunits. (<b>B</b>) MBP-tagged gp3 is co-expressed with gp2 in <span class="html-italic">E. coli</span>. The complex is isolated on amylose beads, MBP tag is cleaved and the complex subsequently purified by size exclusion chromatography using a Superdex 200 column, which yields two peaks. Peak 2 is consistent with ~300–350 kDa, whereas peak 1 is larger (1 MDa–700 kDa). Peaks 1 and 2 are indistinguishable in SDS gel analysis and are initially in dynamic equilibrium. Peak 2 was run three times over gel filtration to obtain a single peak (red trace). Peak 2 was used for all studies herein. Two fractions from each peak were analyzed on SDS-Page gels. Large terminase (L-terminase) and small terminase (S-terminase) molecular weights are denoted by * and **, respectively.</p>
Full article ">Figure 3
<p>Focused classification in 2D. (<b>A</b>) Class averages representing various assembly states after reference-based alignment with SPIDER [<a href="#B40-pathogens-13-01066" class="html-bibr">40</a>]. Masked regions for focused classification are circled in red. Putative gp3 and gp2 regions are denoted with blue and green arrows, respectively. Ellipsoid gp2 region denoted with a green arrow. (<b>B</b>) Resultant 2D class averages with RELION after masking. (<b>C</b>) The 9-mer S-terminase density strongly resembles the P22 gp3 crystal structure with C-terminal helices modeled (PDB 3P9A), giving confidence in focused classification results. The 2D density shows S-terminase (blue) with two gp2s (purple and green) attached at S-terminase C-terminus. The full-length gp2 homolog is from Sf6 (PDB 4IEE) [<a href="#B25-pathogens-13-01066" class="html-bibr">25</a>].</p>
Full article ">Figure 4
<p>S-terminase (gp3)<sub>9</sub> attached to an L-terminase (gp2) core. (<b>A</b>) Class average matches P22 terminase holoenzyme complex containing 1 (gp3)<sub>9</sub> (cyan) subunit and 2 gp2 (ATPase-red and nuclease yellow) subunits (EMD-6429). (<b>B</b>) Iterative Stable Alignment and Classification (ISAC) 2D class averages. Class averages contained one, two, or three (gp3)<sub>9</sub> subunits (blue arrow) attached to a gp2 core (red arrow). Scale bar is 100 Å. (<b>C</b>) Class averages are from (<b>B</b>). Gp3 residues 1–139 (purple) manually docked into representative gp3 density.</p>
Full article ">Figure 5
<p>Mass spectrometric analysis of purified gp3:gp2 complex. (<b>A</b>) The differential assembly is consistent with at least two distinct gp3:gp2 populations of ~290 kDa (stoichiometry (1 × gp3) + (2 × gp2:AMP-PNP)) and ~642 kDa (stoichiometry (2 × gp3) + (5 × gp2:AMP-PNP)) and a ~925 kDa oligomer of the first two complexes combined (stoichiometry (3 × gp3) + (7 × gp2:AMP-PNP)). (<b>B</b>) Representative class averages of the three major assemblies identified by mass spec.</p>
Full article ">Figure 6
<p>Generation of procapsid–terminase complex. Native agarose gel with procapsid (P) incubated with terminase complex (T) forms a new complex (P/T) in the presence of glutaraldehyde crosslinking. Lane 1 contains 0.01% glutaraldehyde and lanes 4–6 contain 0.1% glutaraldehyde and are replicates of the same experiment. Terminase and procapsid alone are shown in lanes 7 and 8, respectively. The crosslinked procapsid–terminase complex appears in lane 1 denoted as P/T complex (yellow dashed rectangle).</p>
Full article ">
17 pages, 693 KiB  
Article
Prevalence of ApoE Alleles in a Spanish Population of Patients with a Clinical Diagnosis of Alzheimer’s Disease: An Observational Case-Control Study
by Laura Bello-Corral, Jesús Seco-Calvo, Angela Molina Fresno, Ana Isabel González, Ana Llorente, Diego Fernández-Lázaro and Leticia Sánchez-Valdeón
Medicina 2024, 60(12), 1941; https://doi.org/10.3390/medicina60121941 - 25 Nov 2024
Viewed by 607
Abstract
Background and Objectives: Alzheimer’s dementia is a progressive neurodegenerative disease that affects memory abilities due to genetic and environmental factors. A well-known gene that influences the risk of Alzheimer’s disease is the apolipoprotein E (APOE) gene. The APOE gene is involved in [...] Read more.
Background and Objectives: Alzheimer’s dementia is a progressive neurodegenerative disease that affects memory abilities due to genetic and environmental factors. A well-known gene that influences the risk of Alzheimer’s disease is the apolipoprotein E (APOE) gene. The APOE gene is involved in the production of a protein that helps transport cholesterol and other types of fat in the bloodstream. Problems in this process are thought to contribute to the development of Alzheimer’s disease. APOE comes in several forms, which are called alleles (ε2, ε3, ε4). Materials and Methods: Therefore, our study aims to identify those subjects with a higher genetic risk through the polymorphism of the APOE gene, using a population screening in patients with a clinical diagnosis of AD in a region of Spain, Castilla y León, as potential biomarkers and to identify individuals at increased genetic risk by polymorphism of the APOE gene. An observational case-control study was conducted in Castilla y León (Spain). Saliva samples were collected and the ApoE gene was analyzed by PCR and agarose gel electrophoresis, respecting ethical criteria. Results: In the Alzheimer’s population in Castilla y León, a high prevalence of ApoE3 (74%) was found, followed by ApoE4 (22%); in addition, a higher presence of the ε4 allele was found in the Alzheimer’s disease (AD) group than in the control group. It was also observed that the ε2/ε2 genotype was not found in any individual with AD but was found in healthy subjects and that the opposite was observed for the ε4/ε4 genotype. The odds ratio (OR) indicated a risk four times greater of having AD if having the ε4 allele. Conclusions: The demonstrated relation between the different isoforms and the likelihood of developing AD has led to its consideration as a biomarker and a potential pre-symptomatic therapy. The molecular mechanisms that confer a disruptive and protective role to ApoE4 and ApoE2, respectively, are still being studied. Full article
(This article belongs to the Section Genetics and Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Distribution of genotypes per group.</p>
Full article ">
20 pages, 4998 KiB  
Article
Bio-Inspired Polymeric Solid Lipid Nanoparticles for siRNA Delivery: Cytotoxicity and Cellular Uptake In Vitro
by Keelan Jagaran, Saffiya Habib and Moganavelli Singh
Polymers 2024, 16(23), 3265; https://doi.org/10.3390/polym16233265 - 24 Nov 2024
Viewed by 570
Abstract
Nanomedicine has introduced strategies that provide precise diagnosis and treatment with fewer side effects than traditional therapies. Treatments for neurodegenerative disorders, including Parkinson’s disease, are palliative, necessitating an innovative delivery system with a curative function. This study investigated a solid lipid nanoparticle (SLNP) [...] Read more.
Nanomedicine has introduced strategies that provide precise diagnosis and treatment with fewer side effects than traditional therapies. Treatments for neurodegenerative disorders, including Parkinson’s disease, are palliative, necessitating an innovative delivery system with a curative function. This study investigated a solid lipid nanoparticle (SLNP) system’s ability to bind and safely deliver siRNA in vitro. SLNPS were formulated using sphingomyelin and cholesterol, with Ginkgo biloba leaf extract (GBE) incorporated to enhance biocompatibility and neuroprotection. Poly-L-lysine (PLL) functionalization ensured successful siRNA binding, safe transport, and protection from nuclease degradation. SLNPs were physicochemically characterized, with binding and protection of siRNA assessed using agarose gels. Cytotoxicity, apoptotic induction, and cellular uptake studies were undertaken in the human neuroblastoma (SH-SY5Y) and embryonic kidney (HEK293) cells. The GBE-PLL-SLNPs had an average size of 93.2 nm and demonstrated enhanced binding and protection of the siRNA from enzyme digestion, with minimal cytotoxicity in HEK293 (<10%) and SH-SY5Y cells (<15%). Caspase 3/7 activity was significantly reduced in both cells, while efficient cellular uptake was noted. The present study provided a solid basis as a proof of principle study for future applications of the potential therapeutic in vitro, promising to address the unmet medical needs of patients with neurological disorders. Full article
(This article belongs to the Section Polymer Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>UV–vis spectra of GBE-SLNPs, H<sub>2</sub>O-SLNPs, GBE-PLL-SLNPs, and H<sub>2</sub>O-PLL-SLNPs. The amplified 320–420 nm section shows the peak and trough variations that are indicated by the arrows.</p>
Full article ">Figure 2
<p>FTIR spectra of: (<b>A</b>) GBE, (<b>B</b>) GBE-SLNPs, (<b>C</b>) GBE-PLL-SLNPs, (<b>D</b>) H<sub>2</sub>O-SLNPs, and (<b>E</b>) H<sub>2</sub>O-PLL-SLNPs.</p>
Full article ">Figure 3
<p>TEM micrographs of the SLNPs. (<b>A</b>) H<sub>2</sub>O-SLNPs, (<b>B</b>) H<sub>2</sub>O-PLL-SLNPs, (<b>C</b>) GBE-SLNPs, (<b>D</b>) GBE-PLL-SLNPs. Scale bar = 200 nm.</p>
Full article ">Figure 4
<p>Ethidium bromide intercalation assay of (<b>A</b>) GBE- and H<sub>2</sub>O-PLL-SLNPs and (<b>B</b>) GBE- and H<sub>2</sub>O-SLNPs with siRNA (0.3 μg/μL). Arrows indicate the inflection points in (<b>A</b>).</p>
Full article ">Figure 5
<p>(<b>A</b>) The band shift assay. (<b>a</b>) GBE-PLL-SLNPs: Lanes 1–8 (0, 0.2, 0.4, 0.6, 0.8, 0.10, 0.12, 0.14 µg) and (<b>b</b>) H<sub>2</sub>O-PLL-SLNPs: Lanes 1–8 (0, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7 µg). The siRNA was maintained at 0.5 µg. The red boxes indicate the optimum binding ratios, superseded by the supra-optimum ratio and preceded by the sub-optimum ratio. (<b>B</b>) The RNase protection assay. In both (<b>a</b>,<b>b</b>), Lanes 1 and 2 contain the positive (undigested siRNA) and negative (digested siRNA) controls. (<b>a</b>) GBE-PLL-SLNPs: Lane 3–5 (0.2, 0.4, 0.6 µg) and (<b>b</b>) H<sub>2</sub>O-PLL-SLNPs: Lanes 3–5 (0.1, 0.2, 0.3 µg). All nanocomplexes were complexed with the targeted siRNA (0.5 µg).</p>
Full article ">Figure 6
<p>The MTT cytotoxicity assay in (<b>A</b>) HEK293 and (<b>B</b>) SH-SY5Y cells. Data are presented as means ± standard deviation (<span class="html-italic">n</span> = 3). A significant difference was observed between the treated cells and the control group (*** <span class="html-italic">p</span> &lt; 0.001; Tukey’s multiple comparisons test).</p>
Full article ">Figure 7
<p>Caspase 3/7 activity induced by the nanocomplexes at the three studied ratios in the (<b>A</b>) HEK293 and (<b>B</b>) SH-SHY5Y cells. The cytographs depict the apoptotic responses of the (<b>B</b>) cells following treatment with the PLL-SLNPs at the sub-optimum, optimum, and supra-optimum ratios.</p>
Full article ">Figure 8
<p>(<b>A</b>) HEK293 cells and (<b>B</b>) SH-SY5Y cells treated with BLOCK-iT™ fluorescent oligo (0.5 and 1.0 µg) conjugated to PLL-SLNPs at 1:1 and 2:1 (<span class="html-italic">w</span>/<span class="html-italic">w</span>) ratios. Intracellular fluorescence was measured after 24 h. (<b>A</b>) Control 1 = HEK293 cells only, and control 2 = naked/uncomplexed BLOCK-iT™ fluorescent oligo. Controls were compared to the nanocomplexes (*** <span class="html-italic">p</span> &lt; 0.001) and further compared with the two treatment groups as indicated by the dotted line (** <span class="html-italic">p</span> &lt; 0.005). (<b>B</b>) Control 1 = SH-SY5Y cells only, and control 2 = naked BLOCK-iT™ fluorescent oligo. Controls were compared to the nanocomplexes and further compared with the two treatment groups as indicated by the dotted line (*** <span class="html-italic">p</span> &lt; 0.001). Statistically significant differences in the therapeutics were noted in both (<b>A</b>,<b>B</b>).</p>
Full article ">
10 pages, 1087 KiB  
Article
Co-Extraction of DNA and RNA from Candida albicans Using a Chemical Method in Conjunction with Silicon Carbide with Few Cells
by Elizabeth Cristina Vieira de Freitas, Francisca Alves dos Santos, Maria Raíssa Vieira Lopes, Dárcio Luiz de Sousa Júnior, Tássia Thaís Al Yafawi, Ana Carolina Ferreira Araújo, Priscilla Ramos Freitas, Irwin Rose Alencar de Menezes, Henrique Douglas Melo Coutinho and Maria Karollyna do Nascimento Silva Leandro
DNA 2024, 4(4), 417-426; https://doi.org/10.3390/dna4040029 - 12 Nov 2024
Viewed by 606
Abstract
Objective: The study aimed to optimize protocols for the joint extraction of deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) from 0.025 × 106 CFU of Candida albicans, targeting to overcome the challenges in the extraction of these genetic materials. Materials and [...] Read more.
Objective: The study aimed to optimize protocols for the joint extraction of deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) from 0.025 × 106 CFU of Candida albicans, targeting to overcome the challenges in the extraction of these genetic materials. Materials and methods: From this, treated silicon carbide (SiC) granules were added to fungal samples from methods 1, 2, and 3 obtained from aliquots of BHI or Sabouraud medium to cause cell lysis and enable the isolation of these macromolecules by phenol and chloroform. The concentration and integrity of the extracted nucleic acids were analyzed, respectively, by spectrophotometry using the A260/A280 ratios and 1% agarose gel electrophoresis. Results: Therefore, method 3 is the one that most comprises samples considered pure of both DNA and RNA, simultaneously. Furthermore, the presence of intact RNAs corresponding to the base pair size such as 5.8 S rRNA and tRNA was verified during electrophoresis, considering the particularities of RNA, which makes it very unstable and easily degraded. Conclusions: Thus, it results in a faster and simpler method in addition to obtain promising results using minimal amounts of biological sample and offering a valuable alternative for small laboratories to work with molecular biology. Full article
Show Figures

Figure 1

Figure 1
<p>Nucleic acid integrity analysis in 1% agarose gel. M: DNA Ladder 1 kb Plus—exACTGene Fischer BioReagent; (<b>A</b>) samples 1 to 10, method 1; (<b>B</b>) method 2; (<b>C</b>) method 3.</p>
Full article ">Scheme 1
<p>Nucleic acid purification and precipitation process. Source: Prepared by the author on the Biorender platform.</p>
Full article ">
13 pages, 3380 KiB  
Article
A Loop-Mediated Isothermal Amplification Assay Utilizing Hydroxy Naphthol Blue (LAMP-HNB) for the Detection of Treponema pallidum Subspp. pallidum
by Saranthum Phurijaruyangkun, Pongbun Tangjitrungrot, Pornpun Jaratsing, Suphitcha Augkarawaritsawong, Khurawan Kumkrong, Sawanya Pongparit, Pawita Suwanvattana, Supatra Areekit, Kosum Chansiri and Somchai Santiwatanakul
Pathogens 2024, 13(11), 949; https://doi.org/10.3390/pathogens13110949 - 31 Oct 2024
Viewed by 763
Abstract
Treponema pallidum subspp. pallidum is a spirochaete bacterium that causes syphilis, one of the most common sexually transmitted diseases. Syphilis progresses through four distinct stages, each characterized by specific symptoms, namely primary, secondary, latent, and late (tertiary) syphilis. Serology has been considered the [...] Read more.
Treponema pallidum subspp. pallidum is a spirochaete bacterium that causes syphilis, one of the most common sexually transmitted diseases. Syphilis progresses through four distinct stages, each characterized by specific symptoms, namely primary, secondary, latent, and late (tertiary) syphilis. Serology has been considered the primary diagnostic approach. However, it is plagued by problems such as the limited specificity of nontreponemal tests and the inadequate correlation of treponemal tests with disease activity. In this study, we focused on the development of a loop-mediated isothermal amplification assay utilizing hydroxy naphthol blue (LAMP-HNB) for the diagnosis of T. pallidum subspp. pallidum. Specifically, this study seeks to determine the analytical sensitivity (limit of detection; LOD) and analytical specificity. Four hundred clinical serum samples were analyzed for diagnostic sensitivity, specificity, and predictive value, and each technique’s 95% confidence intervals (95% CI, p < 0.05) were evaluated. The limit of detection for polymerase chain reaction with agarose gel electrophoresis (PCR-AGE), the loop-mediated isothermal amplification assay combined with agarose gel electrophoresis (LAMP-AGE), and LAMP-HNB were 116 pg/µL, 11.6 pg/µL, and 11.6 pg/ µL, respectively. Analytical specificity examinations indicated the absence of cross-reactivity with Leptospira interrogans, Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, human immunodeficiency virus (HIV), and healthy human serum in PCR-AGE, LAMP-AGE, and LAMP-HNB. The diagnostic sensitivity, diagnostic specificity, positive predictive value (PPV), and negative predictive value (NPV) for PCR-AGE were 100.00 (100.00)%, 94.50 (94.40–94.60)%, 94.79 (94.69–94.88)%, and 100.00 (100.00)%, respectively. While, for LAMP-AGE and LAMP-HNB, they were 100.00 (100.00)%, 91.00 (90.87–91.13)%, 91.74 (91.63–91.86)%, and 100.00 (100.00)%, respectively. The LAMP-HNB test is simple, rapid, highly sensitive, and highly specific, without requiring expensive equipment. In the future, the LAMP-HNB assay may develop into a single-step diagnostic process, enabling the use as point-of-care testing for the diagnosis, prevention, and management of syphilis infection. Full article
Show Figures

Figure 1

Figure 1
<p>The change in the color of hydroxy naphthol blue (HNB) is attributed to the decrease in magnesium. The concentration of Mg<sup>2+</sup> in the solution decreases during the LAMP process, the color of the HNB solution changes from purple to blue, which can be observed with the naked eye after amplification.</p>
Full article ">Figure 2
<p>The optimization of the LAMP assay in the range of 4.5–5.5 mM. Lane “M” represents a 100 bp plus DNA ladder marker of Vivantis, Darul Ehsan, Malaysia, and “N” is the negative control.</p>
Full article ">Figure 3
<p>The optimization of thermal conditions ranging between 60 and 65 °C. Lane “M” represents a 100 bp plus DNA ladder marker of Vivantis, Darul Ehsan, Malaysia, and “Neg” is the negative control.</p>
Full article ">Figure 4
<p>The optimal reaction durations were observed at 45 and 60 min. Lane “M” represents a 100 bp plus DNA ladder marker of Vivantis, Darul Ehsan, Malaysia, and “Neg” is the negative control.</p>
Full article ">Figure 5
<p>The optimization of LAMP-HNB, ranging from 1 to 20 µM.</p>
Full article ">Figure 6
<p>The analytical sensitivity and specificity tests and DNA analysis using 10-fold dilution. In the agarose gel electrophoresis (AGE) results, lanes 1–9: 11.6 ng/µL, 1.16 ng/µL, 116 pg/µL, 11.6 pg/µL, 1.16 pg/µL, 116 fg/µL, 11.6 fg/µL, 1.16 fg/µL, and negative control, respectively. Lane “M” represent a 100 bp plus DNA ladder marker of Vivantis. (<b>a</b>) PCR with agarose gel electrophoresis (PCR-AGE). (<b>b</b>) LAMP with agarose gel electrophoresis (LAMP-AGE). (<b>c</b>) LAMP utilizing hydroxy naphthol blue (LAMP-HNB).</p>
Full article ">Figure 7
<p>In the agarose gel electrophoresis (AGE) results, lanes 1–11: <span class="html-italic">T. pallidum</span> subspp. <span class="html-italic">pallidum</span>, <span class="html-italic">Leptospira interrogans</span>, <span class="html-italic">Staphylococcus aureus</span>, <span class="html-italic">Enterococcus faecalis</span>, <span class="html-italic">Escherichia coli</span>, <span class="html-italic">Klebsiella pneumoniae</span>, <span class="html-italic">Acinetobacter baumannii</span>, <span class="html-italic">Pseudomonas aeruginosa</span>, Human Immunodeficiency Virus (HIV), healthy human serum, and negative control, respectively. Lane “M” represent a 100 bp plus DNA ladder marker of Vivantis. (<b>a</b>) PCR with agarose gel electrophoresis (PCR-AGE). (<b>b</b>) LAMP with agarose gel electrophoresis (LAMP-AGE). (<b>c</b>) LAMP utilizing hydroxy naphthol blue (LAMP-HNB).</p>
Full article ">
Back to TopTop