[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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (644)

Search Parameters:
Keywords = cell penetrating peptide

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
9 pages, 1873 KiB  
Brief Report
Cell-Penetrating Peptide Enhances Tafazzin Gene Therapy in Mouse Model of Barth Syndrome
by Rahul Raghav, Junya Awata, Gregory L. Martin, Douglas Strathdee, Robert M. Blanton and Michael T. Chin
Int. J. Mol. Sci. 2024, 25(24), 13560; https://doi.org/10.3390/ijms252413560 - 18 Dec 2024
Viewed by 286
Abstract
Barth Syndrome (BTHS) is an early onset, lethal X-linked disorder caused by a mutation in tafazzin (TAFAZZIN), a mitochondrial acyltransferase that remodels monolysocardiolipin (MLCL) to mature cardiolipin (CL) and is essential for normal mitochondrial, cardiac, and skeletal muscle function. Current gene therapies in [...] Read more.
Barth Syndrome (BTHS) is an early onset, lethal X-linked disorder caused by a mutation in tafazzin (TAFAZZIN), a mitochondrial acyltransferase that remodels monolysocardiolipin (MLCL) to mature cardiolipin (CL) and is essential for normal mitochondrial, cardiac, and skeletal muscle function. Current gene therapies in preclinical development require high levels of transduction. We tested whether TAFAZZIN gene therapy could be enhanced with the addition of a cell-penetrating peptide, penetratin (Antp). We found that TAFAZZIN-Antp was more effective than TAFAZZIN at preventing the development of pathological cardiac hypertrophy and heart failure. These findings indicate that a cell-penetrating peptide enhances gene therapy for BTHS. Full article
(This article belongs to the Special Issue Exploring Rare Diseases: Genetic, Genomic and Metabolomic Advances)
Show Figures

Figure 1

Figure 1
<p>Schematic presentation of 3rd generation lentivirus production and transduction of TAZ KO mouse embryo fibroblasts (MEFs). (<b>A</b>) Lentiviral construct containing the gene of interest along with the lentiviral packaging plasmids are co-transfected into 293T cells. Following incubation of cells, supernatant containing lentivirus is harvested, purified, and concentrated. (<b>B</b>) MLCL/CL ratio in MEFs after lentivirus treatment. Statistical analysis is performed using IBM SPSS-29 software. One-way ANOVA is conducted between the groups, followed by Bonferonni correction for multiple comparison testing. The results are considered significant when the corrected probability level is &lt;0.05. **** Indicates the significant difference between the Wild/hTAFAZZIN/hTAFAZZIN-Antp and GFP-treated TAFAZZIN KO MEFs at <span class="html-italic">p</span> &lt; 0.0001. Nonsignificant differences are indicated by “ns”.</p>
Full article ">Figure 1 Cont.
<p>Schematic presentation of 3rd generation lentivirus production and transduction of TAZ KO mouse embryo fibroblasts (MEFs). (<b>A</b>) Lentiviral construct containing the gene of interest along with the lentiviral packaging plasmids are co-transfected into 293T cells. Following incubation of cells, supernatant containing lentivirus is harvested, purified, and concentrated. (<b>B</b>) MLCL/CL ratio in MEFs after lentivirus treatment. Statistical analysis is performed using IBM SPSS-29 software. One-way ANOVA is conducted between the groups, followed by Bonferonni correction for multiple comparison testing. The results are considered significant when the corrected probability level is &lt;0.05. **** Indicates the significant difference between the Wild/hTAFAZZIN/hTAFAZZIN-Antp and GFP-treated TAFAZZIN KO MEFs at <span class="html-italic">p</span> &lt; 0.0001. Nonsignificant differences are indicated by “ns”.</p>
Full article ">Figure 2
<p>Tafazzin lentiviral gene therapy affects MLCL/CL ratio, cardiac hypertrophy, and cardiac function. (<b>A</b>) Schematic illustration of methodology adopted for TAFAZZIN KO mice lentivirus treatment: Baseline echocardiograms (ECHOs) were performed on TAFAZZIN KO mice prior to systemic treatment with a single dose of lentivirus (~2.5 × 10<sup>8</sup> transducing units) via tail vein injection at 4 weeks of age (hTAFAZZIN, hTAFAZZIN-Antp, GFP, and empty virus). Serial ECHOs were performed every 4 weeks until 16 weeks. After that, mice were euthanized for tissue harvest and downstream analysis. (<b>B</b>) MLCL/CL ratio in mouse hearts. There is no significant difference between the KO, empty, and GFP-treated mice but a significant difference was found when we compared the KO/GFP/empty-treated mice with the Wild/hTAFAZZIN-Antp/hTAFAZZIN-treated mice (<b>C</b>) Interventricular septum thickness. (<b>D</b>) Left ventricular ejection fraction. (<b>E</b>) Left ventricular fractional shortening. (<b>F</b>) Left ventricular global longitudinal strain. Statistical analysis was performed using IBM SPSS-29 software. One-way ANOVA was conducted between the groups, followed by Bonferonni correction for multiple comparison testing. Within the groups, multiple comparisons were carried out by repeated measures ANOVA with correction by the Bonferroni method. The results are considered significant when the corrected probability level is &lt;0.05 (**/##/@@/<span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001). *** Indicates the significant difference between the wild type and the empty/GFP-treated mice. ## indicates the significant difference between hTAFAZZIN-Antp and empty/GFP-treated mice. In (<b>C</b>,<b>D</b>), @@/<span>$</span><span>$</span> indicates the significant difference between the time points within each group (empty or GFP). (<b>G</b>) Photomicrograph A,B show the GFP immuno-positive cells in the heart section of GFP lentivirus-treated TAFAZZIN KO mouse at 20× and 100× total magnification. Photomicrograph C,D show the negative control (no primary antibody) for GFP at 20× and 100× total magnification.</p>
Full article ">Figure 2 Cont.
<p>Tafazzin lentiviral gene therapy affects MLCL/CL ratio, cardiac hypertrophy, and cardiac function. (<b>A</b>) Schematic illustration of methodology adopted for TAFAZZIN KO mice lentivirus treatment: Baseline echocardiograms (ECHOs) were performed on TAFAZZIN KO mice prior to systemic treatment with a single dose of lentivirus (~2.5 × 10<sup>8</sup> transducing units) via tail vein injection at 4 weeks of age (hTAFAZZIN, hTAFAZZIN-Antp, GFP, and empty virus). Serial ECHOs were performed every 4 weeks until 16 weeks. After that, mice were euthanized for tissue harvest and downstream analysis. (<b>B</b>) MLCL/CL ratio in mouse hearts. There is no significant difference between the KO, empty, and GFP-treated mice but a significant difference was found when we compared the KO/GFP/empty-treated mice with the Wild/hTAFAZZIN-Antp/hTAFAZZIN-treated mice (<b>C</b>) Interventricular septum thickness. (<b>D</b>) Left ventricular ejection fraction. (<b>E</b>) Left ventricular fractional shortening. (<b>F</b>) Left ventricular global longitudinal strain. Statistical analysis was performed using IBM SPSS-29 software. One-way ANOVA was conducted between the groups, followed by Bonferonni correction for multiple comparison testing. Within the groups, multiple comparisons were carried out by repeated measures ANOVA with correction by the Bonferroni method. The results are considered significant when the corrected probability level is &lt;0.05 (**/##/@@/<span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001). *** Indicates the significant difference between the wild type and the empty/GFP-treated mice. ## indicates the significant difference between hTAFAZZIN-Antp and empty/GFP-treated mice. In (<b>C</b>,<b>D</b>), @@/<span>$</span><span>$</span> indicates the significant difference between the time points within each group (empty or GFP). (<b>G</b>) Photomicrograph A,B show the GFP immuno-positive cells in the heart section of GFP lentivirus-treated TAFAZZIN KO mouse at 20× and 100× total magnification. Photomicrograph C,D show the negative control (no primary antibody) for GFP at 20× and 100× total magnification.</p>
Full article ">
11 pages, 2016 KiB  
Article
Fusion Partner Facilitates Expression of Cell-Penetrating Peptide L2 in Pichia pastoris
by Xuan Li, Na Yang, Yuxin Fang, Ruoyu Mao, Ya Hao, Da Teng, Na Dong, Anshan Shan and Jianhua Wang
Antibiotics 2024, 13(12), 1207; https://doi.org/10.3390/antibiotics13121207 - 11 Dec 2024
Viewed by 541
Abstract
Background: L2 is formed by combining the pheromone of Streptococcus agalactiae (S. agalactiae) and a cell-penetrating peptide (CPP) with cell-penetrating selectivity. L2 has more significant penetration and better specificity for killing S. agalactiae. However, the production of AMPs by chemical [...] Read more.
Background: L2 is formed by combining the pheromone of Streptococcus agalactiae (S. agalactiae) and a cell-penetrating peptide (CPP) with cell-penetrating selectivity. L2 has more significant penetration and better specificity for killing S. agalactiae. However, the production of AMPs by chemical synthesis is always a challenge because of the production cost. Methods: This study was devoted to the heterologous expression of the cell-penetrating peptide L2 in Pichia pastoris using SUMO and a short acidic fusion tag as fusion partners, and the high-density expression of SUMO-L2 was achieved in a 5 L fermenter. Results: The results showed that SUMO-L2 expression in the 5 L fermenter reached 629 mg/L. The antibacterial activity of recombinant L2 was examined; the minimum inhibitory concentration (MICs) and minimum bactericidal concentration (MBCs) of purified L2 were 4–8 μg/mL and 8–16 μg/mL against S. agalactiae after 84 h of lysis with 50% formic acid. Conclusions: The findings suggest that SUMO is a suitable fusion tag to express cell-penetrating peptide L2. Full article
(This article belongs to the Special Issue Bioactive Peptides and Their Antibiotic Activity)
Show Figures

Figure 1

Figure 1
<p>Construction and extraction of recombinant plasmids. (<b>a</b>,<b>b</b>) Schematic diagram of recombinant expression plasmids pPIC-SUMO-L2 and pPIC-2FLAG-L2. (<b>c</b>) Gel analysis of the plasmids pPIC-2FLAG-L2 and pPIC-SUMO-L2. Lane M: Trans5K DNA marker; Lanes 1 and 2: unenzymatically cleaved plasmids pPIC-2FLAG-L2 and pPIC-SUMO-L2; Lanes 3 and 4: linearized plasmids of pPIC-2FLAG-L2 and pPIC-SUMO-L2.</p>
Full article ">Figure 2
<p>Large-volume induction of recombinant proteins; (<b>a</b>) Tricine SDS-PAGE to detect the fermentation supernatant after 120 h of horizontal induction in shaking flasks. Lane M: protein marker (6 μL); Lanes 1–3 represent the transformants of SUMO-L2 named 25, 44, 52 (10 μL), and Lanes 4–6 represent the transformants of 2FLAG-L2 named 50, 51, 52 (10 μL). (<b>b</b>) Tricine SDS-PAGE to detect the fermentation supernatant after 72 h of horizontal induction in a 5 L fermenter of SUMO-L2. (<b>c</b>) Changes in total protein concentration and wet weight of <span class="html-italic">P. pastoris</span> with time during high-density fermentation of SUMO-L2.</p>
Full article ">Figure 3
<p>SUMO-L2 purification results by Ni<sup>2+</sup>-chelate affinity column and schematic diagram of formic acid cutting time mapping. (<b>a</b>) Tricine-SDS-PAGE results of purified SUMO-L2. Lane M: protein marker; Lane 1: fermentation of unpurified samples; Lane 2: penetration peaks; Lanes 3–4: decontamination peaks; Lane 5: destination peaks; Lane 6: washed peaks. (<b>b</b>) Tricine-SDS-PAGE results showed that SUMO-L2 was cleaved using formic acid. Lane M: protein marker; and Lanes 1–6: represent the cleavage time of 0 h, 24 h, 48 h, 72 h, 84 h, and 96 h, respectively. (<b>c</b>) Activity test by inhibition zone after formic acid cleavage of SUMO-L2.</p>
Full article ">Figure 4
<p>Purification and identification of L2. (<b>a</b>) Tricine-SDS-PAGE analysis and purification recovery of L2; bands are in the red circle. M: Spectra™ Multicolor Low Range Protein Ladder. Lane 1: SUMO-L2 after cutting by formic acid; Lane 2: purified product. (<b>b</b>) MALDI-TOF MS analysis of purified L2.</p>
Full article ">Figure 5
<p>Modeling the secondary structure of L2 and fusion tags using I-TASSER (<a href="https://zhanggroup.org/I-TASSER/" target="_blank">https://zhanggroup.org/I-TASSER/</a>, accessed on 9 April 2024).</p>
Full article ">
16 pages, 3181 KiB  
Article
Tumor-Targeted Cell-Penetrating Peptides Reveal That Monomethyl Auristatin E Temporally Modulates the Tumor Immune Microenvironment
by Mahsa Mortaja, Marcus M. Cheng, Alina Ali, Jacqueline Lesperance, Dina V. Hingorani, Mike M. Allevato, Kanika Dhawan, Maria F. Camargo, Rana R. McKay, Stephen R. Adams, J. Silvio Gutkind and Sunil J. Advani
Molecules 2024, 29(23), 5618; https://doi.org/10.3390/molecules29235618 - 27 Nov 2024
Viewed by 586
Abstract
Chemotherapies remain standard therapy for cancers but have limited efficacy and cause significant side effects, highlighting the need for targeted approaches. In the progression of cancer, tumors increase matrix metalloproteinase (MMP) activity. Leveraging and therapeutically redirecting tumor MMPs through activatable cell-penetrating peptide (ACPP) [...] Read more.
Chemotherapies remain standard therapy for cancers but have limited efficacy and cause significant side effects, highlighting the need for targeted approaches. In the progression of cancer, tumors increase matrix metalloproteinase (MMP) activity. Leveraging and therapeutically redirecting tumor MMPs through activatable cell-penetrating peptide (ACPP) technology offers new approaches for tumor-selective drug delivery and for studying how drug payloads engage the tumor immune microenvironment. ACPPs are biosensing peptides consisting of a drug-conjugated polycationic cell-penetrating peptide masked by an autoinhibitory polyanionic peptide through an interlinking peptide linker. Since tumors overexpress MMPs, ACPP tumor-targeting is achieved using an MMP cleavable linker. Monomethyl auristatin E (MMAE) is a potent anti-tubulin and common drug payload in antibody drug conjugates; however there are limited pre-clinical studies on how this clinically effective drug modulates the interplay of cancer cells and the immune system. Here, we report the versatility of ACPP conjugates in syngeneic murine cancer models and interrogate how MMAE temporally alters the tumor immune microenvironment. We show that cRGD-ACPP-MMAE preferentially delivered MMAE to tumors in murine models. Targeted cRGD-ACPP-MMAE demonstrated anti-tumor kill activity that activated the innate and adaptive arms of the immune system. Understanding how targeted MMAE engages tumors can optimize MMAE tumor kill activity and inform rational combinations with other cancer therapeutics. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Tissue-targeting cell-penetrating peptides using matrix metalloproteinases. (<b>A</b>) Gelatin zymography of human xenograft and syngeneic murine tumors with adjacent normal murine muscle tissue. Molecular weight marker (MW) with control MMP-2 and MMP-9 standard activities in far-left lanes. (<b>B</b>) Structural representation of cRGD-ACPP–drug conjugate. Polycationic cell-penetrating peptide (+) and autoinhibitory polyanionic peptide (−) connected by an MMP-2/9 sensitive peptide linker (PLGC(Me)AG, C(Me) denoted C’). (<b>C</b>) Schema for activatable cell-penetrating peptide tumor localization. MMP-2/9 cleavage and cathepsin cleavage site indicated by dashed green lines.</p>
Full article ">Figure 2
<p>Chemical structures of MMP-sensitive activatable penetrating peptides. (<b>A</b>) Synthetic scheme for conjugating monomethyl auristatin E to activatable cell-penetrating peptide. MMAE attached to MC-VC-PABC linker reacted with ACPP and cRGD to yield co-targeted cRGD-ACPP-MMAE. (<b>B</b>) Ratiometric ACPP labeled with Cy5 and Cy7 to the polycationic and polyanionic ends, respectively.</p>
Full article ">Figure 3
<p>Biodistribution of MMP-guided activable cell-penetrating peptides in vivo. (<b>A</b>) Structural representations of activatable cell-penetrating peptide–MMAE conjugate. (<b>B</b>) Structural representations of ratiometric activatable cell-penetrating peptide. (<b>C</b>) Mice with HCT116 tumor xenografts injected with cRGD-ACPP-MMAE. Tissues harvested at indicated time points and drug concentration determined. Data plotted as mean ± SEM. (<b>D</b>) Scatter plot of drug concentration in individual tissues at 24 h post-injection. Statistical significance calculated using one-way ANOVA with Tukey’s multiple comparisons test. (<b>E</b>) Mice with syngeneic subcutaneous LL2 or MC38 tumors (white dotted circles indicate bilateral tumor locations) injected IV with ratiometric ACPP. In situ whole-mouse imaging of Cy5 and Cy7 with Cy5:Cy7 emission ratio calculated. Gut auto-fluorescence indicated by pink arrows. Pseudocolor Cy:C7 emission ratio scale bar shown far right.</p>
Full article ">Figure 4
<p>Anti-tumor efficacy of cRGD-ACPP-MMAE. (<b>A</b>) Mice bearing LL2 tumors IV injected with vehicle (untreated) or cRGD-ACPP-MMAE (20 nmoles on days 4 and 6). Individual tumor volumes plotted, left panel. Average tumors volumes on day 18 plotted as mean ± SEM, right panel (n = 10 for untreated, n = 6 for cRGD-ACPP-MMAE). Statistical significance calculated using two-tailed <span class="html-italic">t</span> testing. (<b>B</b>) Mice bearing B16 tumors IV injected with vehicle (untreated) or cRGD-ACPP-MMAE (20 nmoles on days 3 and 5). Individual tumor volumes plotted, left panel. Average tumors volumes on day 19 plotted as mean ± SEM, right panel (n = 10 for untreated, n = 6 for cRGD-ACPP-MMAE). Statistical significance calculated using two-tailed <span class="html-italic">t</span> testing. (<b>C</b>) Mouse body weights of mice from experiments in (<b>A</b>,<b>B</b>), plotted as mean fractional body weight ± SEM.</p>
Full article ">Figure 5
<p>MMAE-induced temporal changes in the tumor immune microenvironment. Mice with B16 tumors treated with IV cRGD-ACPP-MMAE. Tumors harvested at time 48–120 h post-drug injection and analyzed using NanoString nCounter PanCancer Mouse Immune Profiling panel. Heatmap depicts the Z-score of immune signatures for biological pathways (<b>A</b>) and individual immune cell types (<b>B</b>). (<b>C</b>) Individual immune cell type abundance at 96 h after the injection of cRGD-ACPP-MMAE. Data plotted as scatter plot with mean ± SEM. Statistical significance calculated using two-tailed <span class="html-italic">t</span> testing.</p>
Full article ">
19 pages, 1632 KiB  
Article
New Conjugates of Vancomycin with Cell-Penetrating Peptides—Synthesis, Antimicrobial Activity, Cytotoxicity, and BBB Permeability Studies
by Jarosław Ruczyński, Katarzyna Prochera, Natalia Kaźmierczak, Katarzyna Kosznik-Kwaśnicka, Lidia Piechowicz, Piotr Mucha and Piotr Rekowski
Molecules 2024, 29(23), 5519; https://doi.org/10.3390/molecules29235519 - 22 Nov 2024
Viewed by 641
Abstract
Vancomycin (Van) is a glycopeptide antibiotic commonly used as a last resort for treating life-threatening infections caused by multidrug-resistant bacterial strains, such as Staphylococcus aureus and Enterococcus spp. However, its effectiveness is currently limited due to the rapidly increasing number of drug-resistant clinical [...] Read more.
Vancomycin (Van) is a glycopeptide antibiotic commonly used as a last resort for treating life-threatening infections caused by multidrug-resistant bacterial strains, such as Staphylococcus aureus and Enterococcus spp. However, its effectiveness is currently limited due to the rapidly increasing number of drug-resistant clinical strains and its inherent cytotoxicity and poor penetration into cells and specific regions of the body, such as the brain. One of the most promising strategies to enhance its efficacy appears to be the covalent attachment of cell-penetrating peptides (CPPs) to the Van structure. In this study, a series of vancomycin conjugates with CPPs—such as TP10, Tat (47–57), PTD4, and Arg9—were designed and synthesized. These conjugates were tested for antimicrobial activity against four reference strains (Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, and Pseudomonas aeruginosa) and two clinical drug-resistant strains: methicillin-resistant S. aureus and vancomycin-resistant E. faecium. In addition, cytotoxicity tests (using a human fibroblast cell line) and blood–brain barrier (BBB) permeability tests (using a parallel artificial membrane permeability assay—PAMPA-BBB assay) were conducted for selected compounds. Our research demonstrated that conjugation of Van with CPPs, particularly with Tat (47–57), Arg9, or TP10, significantly enhances its antimicrobial activity against Gram-positive bacteria such as S. aureus and Enterococcus spp., reduces its cytotoxicity, and improves its access to brain tissues. We conclude that these findings provide a strong foundation for the design of novel antimicrobial agents effective in treating infections caused by drug-resistant staphylococcal and enterococcal strains, while also being capable of crossing the BBB. Full article
(This article belongs to the Special Issue Chemical Biology of Antimicrobial Resistance, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the chemical structure of the synthesized Van-CPP conjugates, where <span class="html-italic">V</span><sub>V</sub> is the primary amino position in the carbohydrate moiety of vancomycin (green circle, position of PEG<sub>4</sub> or Suc-Cystamine linker attachment), <span class="html-italic">V</span><sub>C</sub> is the <span class="html-italic">C</span>-terminal carboxylic position of vancomycin (orange circle, position of PEG<sub>3</sub> or Cystamine linker attachment), PEG<sub>4</sub> is 4,7,10,13-tetraoxopentadecane-1-caboxylate, PEG<sub>3</sub> is 1-amino-3,6,9-trioxoundecane, Cystamine is 1-amino-2-(ethyldisulfanyl)ethan, Suc-Cystamine is 4-(2-(ethyldisulfanyl)ethylamino)-4-oxobutane-1-carboxylate, CPP is the <span class="html-italic">N</span>-terminal amino position of cell-penetrating peptides (blue circle, position of CPP attachment): Tat (47–57) and PTD4 (attached via PEG<sub>3</sub>, PEG<sub>4</sub>, Cystamine and Suc-Cystamine linker), Arg<sub>9</sub> (attached via PEG<sub>3</sub>, PEG<sub>4</sub> and Cystamine linker) or TP10 (attached via PEG<sub>3</sub> and PEG<sub>4</sub> linker).</p>
Full article ">Figure 2
<p>Effects of vancomycin conjugates and their components on the viability of normal human fibroblast cells: (<b>A</b>) TP10 and its conjugates, (<b>B</b>) Tat(47–57) and its conjugates, (<b>C</b>) PTD4 and its conjugates, (<b>D</b>) Arg<sub>9</sub> and its conjugates. Cells were incubated with various concentrations of peptides for 24 h and cell viability was assessed by neutral red cytotoxicity assay. Plots present mean ± SD from three independent experiments performed in triplicate. The x-axis represents peptide concentration in µg/mL. The y-axis represents cell viability expressed as a percentage relative to the untreated control cells incubated without peptides as well as control cells treated with 10% DMSO. A one-way ANOVA test was used to test the degree of significance. * statistically significant (<span class="html-italic">p</span> &lt; 0.05) as compared to control.</p>
Full article ">Figure 3
<p>The general scheme of the synthesis of vancomycin conjugates with CPPs showing the preparation of one of the representatives of the conjugates—Van-PEG<span class="html-italic"><sub>3</sub></span>-Tat (47–57). The syntheses consist of three main steps. (<b>I</b>) In the first step, azido-functionalized Van derivatives are obtained by coupling vancomycin with bifunctional linkers, e.g., H<span class="html-italic"><sub>2</sub></span>N-PEG<span class="html-italic"><sub>3</sub></span>-N<span class="html-italic"><sub>3</sub></span> (1-amino-11-azido-3,6,9-trioxoundecane); (<b>II</b>) the second step is the solid-phase synthesis (SPPS) of CPP derivatives with a propiolate group attached to the <span class="html-italic">N</span>-terminus, e.g., Prop-Tat (47–57); (<b>III</b>) in the third step, vancomycin derivatives are conjugated with CPP derivatives using the highly effective and chemoselective 1,3-dipolar Huisgen cycloaddition, commonly known as the “click reaction”, which leads to the formation of a 1,2,3-triazole ring and stable Van-CPP conjugates, e.g., Van-PEG<span class="html-italic"><sub>3</sub></span>-Tat (47–57).</p>
Full article ">
15 pages, 3230 KiB  
Article
Enhanced Ocular Bioavailability and Prolonged Duration via Hydrophilic Surface Nanocomposite Vesicles for Topical Drug Administration
by Sa Huang, Yuan Xu, Yingyao Luo, Zhijiong Wang, Fan Li, Zhenmiao Qin and Junfeng Ban
Pharmaceutics 2024, 16(12), 1496; https://doi.org/10.3390/pharmaceutics16121496 - 21 Nov 2024
Viewed by 418
Abstract
Background: Internal ocular diseases, such as macular edema, uveitis, and diabetic macular edema require precise delivery of therapeutic agents to specific regions within the eye. However, the eye’s complex anatomical structure and physiological barriers present significant challenges to drug penetration and distribution. Traditional [...] Read more.
Background: Internal ocular diseases, such as macular edema, uveitis, and diabetic macular edema require precise delivery of therapeutic agents to specific regions within the eye. However, the eye’s complex anatomical structure and physiological barriers present significant challenges to drug penetration and distribution. Traditional eye drops suffer from low bioavailability primarily due to rapid clearance mechanisms. Methods: The novel ocular drug delivery system developed in this study utilizes poly(lactic-co-glycolic acid) (PLGA) nanoparticles modified with cell-penetrating peptides (CPPs). In vitro drug release studies were conducted to evaluate the sustained-release properties of the nanoparticles. Ex vivo experiments using MDCK cells assessed corneal permeability and uptake efficiency. Additionally, in vivo studies were performed in rabbit eyes to determine the nanoparticles’ resistance to elimination by tears and their retention time in the aqueous humor. Results: In vitro drug release studies demonstrated superior sustained-release properties of the nanoparticles. Ex vivo experiments revealed enhanced corneal permeability and increased uptake efficiency by MDCK cells. In vivo studies in rabbit eyes confirmed the nanoparticles’ resistance to elimination by lacrimal fluid and their ability to extend retention time in the aqueous humor. CPP modification significantly improved ocular retention, corneal penetration, and cellular endocytosis efficiency. Conclusions: The CPP-modified PLGA nanoparticles provide an effective and innovative solution for ocular drug delivery, offering improved bioavailability, prolonged retention, and enhanced drug penetration, thereby overcoming the challenges of traditional intraocular drug administration methods. Full article
(This article belongs to the Special Issue Polymer-Based Delivery System)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Particle size, potential, and microscopic images of NPs. (<b>A</b>) The particle size and potential of NPs. (<b>B</b>) Microscopic image of the NPs.</p>
Full article ">Figure 2
<p>In vitro release and ex vivo corneal permeation studies. (<b>A</b>) The cumulative release rate of NPs. (<b>B</b>) In vitro corneal permeation of NPs.</p>
Full article ">Figure 3
<p>Tear elimination and aqueous humor dynamics studies. (<b>A</b>) TA elimination curve in tears. (<b>B</b>) The pharmacokinetic curves of TA within aqueous humor.</p>
Full article ">Figure 4
<p>Properties and mechanisms of NPs transport across cellular barriers The data are given as mean ± SD. (<b>A</b>) The cytotoxicity study of coumarin-6 nanoparticles. (<b>B</b>) The difference in NP uptake by MDCK cells. (<b>C</b>) Epithelial barrier penetration studies of NPs. (<b>D</b>) The mechanism analysis of endocytosis of MDCK cells monolayer for nanoparticles with different properties by adding various inhibitors. (<b>E</b>) The mechanism analysis of NP transport across the epithelial cell barrier by adding different inhibitors.</p>
Full article ">
56 pages, 5775 KiB  
Review
Gold Nanoparticles in Nanomedicine: Unique Properties and Therapeutic Potential
by Furkan Eker, Emir Akdaşçi, Hatice Duman, Mikhael Bechelany and Sercan Karav
Nanomaterials 2024, 14(22), 1854; https://doi.org/10.3390/nano14221854 - 20 Nov 2024
Viewed by 1518
Abstract
Gold nanoparticles (NPs) have demonstrated significance in several important fields, including drug delivery and anticancer research, due to their unique properties. Gold NPs possess significant optical characteristics that enhance their application in biosensor development for diagnosis, in photothermal and photodynamic therapies for anticancer [...] Read more.
Gold nanoparticles (NPs) have demonstrated significance in several important fields, including drug delivery and anticancer research, due to their unique properties. Gold NPs possess significant optical characteristics that enhance their application in biosensor development for diagnosis, in photothermal and photodynamic therapies for anticancer treatment, and in targeted drug delivery and bioimaging. The broad surface modification possibilities of gold NPs have been utilized in the delivery of various molecules, including nucleic acids, drugs, and proteins. Moreover, gold NPs possess strong localized surface plasmon resonance (LSPR) properties, facilitating their use in surface-enhanced Raman scattering for precise and efficient biomolecule detection. These optical properties are extensively utilized in anticancer research. Both photothermal and photodynamic therapies show significant results in anticancer treatments using gold NPs. Additionally, the properties of gold NPs demonstrate potential in other biological areas, particularly in antimicrobial activity. In addition to delivering antigens, peptides, and antibiotics to enhance antimicrobial activity, gold NPs can penetrate cell membranes and induce apoptosis through various intracellular mechanisms. Among other types of metal NPs, gold NPs show more tolerable toxicity capacity, supporting their application in wide-ranging areas. Gold NPs hold a special position in nanomaterial research, offering limited toxicity and unique properties. This review aims to address recently highlighted applications and the current status of gold NP research and to discuss their future in nanomedicine. Full article
Show Figures

Figure 1

Figure 1
<p>Applications of gold NPs in various fields [<a href="#B3-nanomaterials-14-01854" class="html-bibr">3</a>,<a href="#B5-nanomaterials-14-01854" class="html-bibr">5</a>].</p>
Full article ">Figure 2
<p>Graph representing the published research papers that include “gold nanoparticles” in their title for the last 5 years, with a pie chart showing the distribution of applications based on the discussed sections [<a href="#B8-nanomaterials-14-01854" class="html-bibr">8</a>].</p>
Full article ">Figure 3
<p>General properties of gold nanoparticles [<a href="#B13-nanomaterials-14-01854" class="html-bibr">13</a>].</p>
Full article ">Figure 4
<p>Drug delivery application of gold NPs [<a href="#B29-nanomaterials-14-01854" class="html-bibr">29</a>].</p>
Full article ">Figure 5
<p>Nucleic acid delivery mechanism of gold NPs. Through endocytosis, functionalized gold NPs effectively transport nucleic acids into cells, and surface alterations improve targeting. The nucleic acids are released into the cytoplasm by endosomal escape mechanisms after internalization, providing opportunities for immunotherapy and gene therapy [<a href="#B61-nanomaterials-14-01854" class="html-bibr">61</a>].</p>
Full article ">Figure 6
<p>Representation of protein delivery mechanism of gold NPs. By altering their surfaces with ligands, polymers, or linkers, gold NPs may be made to bind particular proteins. This increases their circu-lation time and stops enzymatic breakdown. Through endocytosis, gold NPs enable cellular ab-sorption and release protein cargo inside cells. Therapeutic applications benefit from surface changes that improve targeting to certain tissues or cell types [<a href="#B75-nanomaterials-14-01854" class="html-bibr">75</a>].</p>
Full article ">Figure 7
<p>Gold NP-based photothermal and photodynamic therapy in anticancer application [<a href="#B52-nanomaterials-14-01854" class="html-bibr">52</a>,<a href="#B129-nanomaterials-14-01854" class="html-bibr">129</a>].</p>
Full article ">Figure 8
<p>Antibacterial activity of gold NPs by multiple mechanisms [<a href="#B209-nanomaterials-14-01854" class="html-bibr">209</a>].</p>
Full article ">Figure 9
<p>Potential toxicity mechanisms of gold NPs [<a href="#B257-nanomaterials-14-01854" class="html-bibr">257</a>,<a href="#B258-nanomaterials-14-01854" class="html-bibr">258</a>].</p>
Full article ">Figure 10
<p>Number of registered patents containing “Gold Nanoparticle” in their title in the last five years [<a href="#B277-nanomaterials-14-01854" class="html-bibr">277</a>].</p>
Full article ">
14 pages, 26460 KiB  
Article
TF-BAPred: A Universal Bioactive Peptide Predictor Integrating Multiple Feature Representations
by Zhenming Wu, Xiaoyu Guo, Yangyang Sun, Xiaoquan Su and Jin Zhao
Mathematics 2024, 12(22), 3618; https://doi.org/10.3390/math12223618 - 20 Nov 2024
Viewed by 419
Abstract
Bioactive peptides play essential roles in various biological processes and hold significant therapeutic potential. However, predicting the functions of these peptides is challenging due to their diversity and complexity. Here, we develop TF-BAPred, a framework for universal peptide prediction incorporating multiple feature representations. [...] Read more.
Bioactive peptides play essential roles in various biological processes and hold significant therapeutic potential. However, predicting the functions of these peptides is challenging due to their diversity and complexity. Here, we develop TF-BAPred, a framework for universal peptide prediction incorporating multiple feature representations. TF-BAPred feeds original peptide sequences into three parallel modules: a novel feature proposed in this study called FVG extracts the global features of each peptide sequence; an automatic feature recognition module based on a temporal convolutional network extracts the temporal features; and a module integrates multiple widely used features such as AAC, DPC, BPF, RSM, and CKSAAGP. In particular, FVG constructs a fixed-size vector graph to represent the global pattern by capturing the topological structure between amino acids. We evaluated the performance of TF-BAPred and other peptide predictors on different types of peptides, including anticancer peptides, antimicrobial peptides, and cell-penetrating peptides. The benchmarking tests demonstrate that TF-BAPred displays strong generalization and robustness in predicting various types of peptide sequences, highlighting its potential for applications in biomedical engineering. Full article
(This article belongs to the Special Issue Bioinformatics and Mathematical Modelling)
Show Figures

Figure 1

Figure 1
<p>Overview of the TF-BAPred framework. (<b>A</b>) The original peptide sequences are individually input into three channels to extract sequence features from different perspectives. Subsequently, the feature vectors obtained from the three channels are fused and input into a fully connected neural network for classification training and prediction. (<b>B</b>) An example of a fixed-scale vector graph depicting the global structural patterns of each peptide sequence. (<b>C</b>) The framework for temporal feature extraction based on a temporal convolutional network.</p>
Full article ">Figure 2
<p>Example of constructing a fixed-scale vector graph. (<b>a</b>) A peptide sequence <math display="inline"><semantics> <mrow> <mi>S</mi> <mo>=</mo> <mi>DVADVMYYV</mi> </mrow> </semantics></math>. (<b>b</b>) Assuming the amino acid alphabet consists of A, D, M, V, and Y, construct a vector graph based on the alphabet. (<b>c</b>) Generate a feature matrix based on the constructed vector graph.</p>
Full article ">Figure 3
<p>ROC curves and corresponding AUC values of the TCN and other compared methods on (<b>A</b>) ACP740, (<b>B</b>) ACPmain, (<b>C</b>) Veltri’s dataset, (<b>D</b>) Ma’s dataset, (<b>E</b>) CPP924, and (<b>F</b>) CPPsite3 datasets.</p>
Full article ">Figure 4
<p>Evaluation of TF-BAPred’s generalizability on anticancer peptide datasets of (<b>A</b>) ACP740 and (<b>B</b>) ACPmain, antimicrobial peptide datasets of (<b>C</b>) Veltri’s dataset and (<b>D</b>) Ma’s dataset, and cell-penetrating peptide datasets of (<b>E</b>) CPP924 and (<b>F</b>) CPPsite3.</p>
Full article ">Figure 5
<p>The accuracies of peptide predictors across different ratios of training and testing datasets on the ACP740 dataset.</p>
Full article ">
17 pages, 3755 KiB  
Article
Immunomodulatory Effects of the Tobacco Defensin NaD1
by Ekaterina I. Finkina, Ivan V. Bogdanov, Olga V. Shevchenko, Serafima I. Fateeva, Anastasia A. Ignatova, Sergey V. Balandin and Tatiana V. Ovchinnikova
Antibiotics 2024, 13(11), 1101; https://doi.org/10.3390/antibiotics13111101 - 19 Nov 2024
Viewed by 612
Abstract
Background/Objectives: Defensins are important components of the innate plant immune system, exhibiting antimicrobial activity against phytopathogens, as well as against fungi pathogenic to humans. Along with antifungal activity, plant defensins are also capable of influencing various immune processes, but not much is known [...] Read more.
Background/Objectives: Defensins are important components of the innate plant immune system, exhibiting antimicrobial activity against phytopathogens, as well as against fungi pathogenic to humans. Along with antifungal activity, plant defensins are also capable of influencing various immune processes, but not much is known about these effects. In this study, we investigated the immunomodulatory effects of the tobacco defensin NaD1, which possesses a pronounced antifungal activity. Methods and Results: We showed that NaD1 could penetrate the Caco-2 polarized monolayer. Using a multiplex assay with a panel of 48 cytokines, chemokines and growth factors, we demonstrated that NaD1 at a concentration of 2 μM had immunomodulatory effects on human dendritic cells and blood monocytes, mainly inhibiting the production of various immune factors. Using the sandwich ELISA method, we demonstrated that NaD1 at the same concentration had a pronounced immunomodulatory effect on unstimulated THP-1-derived macrophages and those stimulated by bacterial LPS or fungal zymosan. NaD1 had a dual effect and induced the production of both pro-inflammatory cytokine IL-1β as well as anti-inflammatory IL-10 on resting and pro-inflammatory THP-1-derived macrophages. We also found that the immunomodulatory effects of the tobacco defensin NaD1 and the pea defensin Psd1 differed from each other, indicating nonuniformity in the modes of action of plant defensins. Conclusions: Thus, our data demonstrated that the tobacco defensin NaD1 exhibits different immunomodulatory effects on various immune cells. We hypothesized that influence on human immune system along with antifungal activity, could determine the effectiveness of this peptide under infection in vivo. Full article
Show Figures

Figure 1

Figure 1
<p>Cytotoxic effects of the tobacco defensin NaD1 towards PBMCs (<b>A</b>) and Caco-2 cells in monolayer (<b>C</b>). The membrane-active peptide melittin from the venom of honeybees (<b>B</b>,<b>D</b>) was used for comparison. Error bars represent a standard deviation (±SD) between two biological and two technical replications. Significance levels are * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001. The significance was calculated by comparing untreated cells (control) with treated by NaD1 or melittin cells. Viability cells in control and experimental samples was compared with un-paired two-sample <span class="html-italic">t</span>-test.</p>
Full article ">Figure 2
<p>Assessment of bidirectional transport of the tobacco defensin NaD1 through the polarized Caco-2 monolayer. A→B, absorptive transport; B→A, secretory transport; Papp—apparent permeability coefficient. Six and four independent biological replications were used for absorptive and secretory directions, respectively. The normality of Papp coefficient distribution was assessed using Shapiro–Wilk test. Papp coefficients were compared by unpaired two-sample <span class="html-italic">t</span>-test.</p>
Full article ">Figure 3
<p>Production of cytokines, chemokines and growth factors upon stimulation of DCs and monocytes by NaD1 at the concentration of 2 μM. Error bars represent a standard deviation (±SD) between two biological replications. The levels in control and experimental wells were compared by unpaired two-sample <span class="html-italic">t</span>-test.</p>
Full article ">Figure 4
<p>Influence of the tobacco defensin NaD1 and other AMPs at the concentration of 2 µM on production of pro- (<b>A</b>–<b>D</b>) and anti-inflammatory (<b>B</b>) cytokines either unstimulated or stimulated by LPS or by zymosan THP-1-derived macrophages. Error bars represent a standard deviation (±SD) between two biological and two technical replications. Significance levels are * <span class="html-italic">p</span> ≤ 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. The significance of difference in cytokine production was calculated by comparing: unstimulated cells (control) with stimulated by AMPs cells (grey bars); stimulated by LPS (blue bars) or zymosan (green bars) cells alone or in the presence of AMPs. Release of the cytokines in control and experimental samples was compared with unpaired two-sample <span class="html-italic">t</span>-test.</p>
Full article ">
21 pages, 4816 KiB  
Article
Roscovitine, a CDK Inhibitor, Reduced Neuronal Toxicity of mHTT by Targeting HTT Phosphorylation at S1181 and S1201 In Vitro
by Hongshuai Liu, Ainsley McCollum, Asvini Krishnaprakash, Yuxiao Ouyang, Tianze Shi, Tamara Ratovitski, Mali Jiang, Wenzhen Duan, Christopher A. Ross and Jing Jin
Int. J. Mol. Sci. 2024, 25(22), 12315; https://doi.org/10.3390/ijms252212315 - 16 Nov 2024
Viewed by 930
Abstract
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disease caused by a single mutation in the huntingtin gene (HTT). Normal HTT has a CAG trinucleotide repeat at its N-terminal within the range of 36. However, once the CAG repeats exceed 37, the mutant [...] Read more.
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disease caused by a single mutation in the huntingtin gene (HTT). Normal HTT has a CAG trinucleotide repeat at its N-terminal within the range of 36. However, once the CAG repeats exceed 37, the mutant gene (mHTT) will encode mutant HTT protein (mHTT), which results in neurodegeneration in the brain, specifically in the striatum and other brain regions. Since the mutation was discovered, there have been many research efforts to understand the mechanism and develop therapeutic strategies to treat HD. HTT is a large protein with many post-translational modification sites (PTMs) and can be modified by phosphorylation, acetylation, methylation, sumoylation, etc. Some modifications reduced mHTT toxicity both in cell and animal models of HD. We aimed to find the known kinase inhibitors that can modulate the toxicity of mHTT. We performed an in vitro kinase assay using HTT peptides, which bear different PTM sites identified by us previously. A total of 368 kinases were screened. Among those kinases, cyclin-dependent kinases (CDKs) affected the serine phosphorylation on the peptides that contain S1181 and S1201 of HTT. We explored the effect of CDK1 and CDK5 on the phosphorylation of these PTMs of HTT and found that CDK5 modified these two serine sites, while CDK5 knockdown reduced the phosphorylation of S1181 and S1201. Modifying these two serine sites altered the neuronal toxicity induced by mHTT. Roscovitine, a CDK inhibitor, reduced the p-S1181 and p-S1201 and had a protective effect against mHTT toxicity. We further investigated the feasibility of the use of roscovitine in HD mice. We confirmed that roscovitine penetrated the mouse brain by IP injection and inhibited CDK5 activity in the brains of HD mice. It is promising to move this study to in vivo for pre-clinical HD treatment. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Kinase assays identified upstream kinases modifying the serine of HTT peptides in vitro. In vitro kinase assay was used to screen the kinase targeting HTT peptide bearing serine 1181, serine 1201, and serine 2653 of HTT. (<b>A</b>) Kinases targeting the serine 1181 of the HTT peptide. (<b>B</b>) Kinases targeting the serine 1201 of the HTT peptide. (<b>C</b>) Kinases targeting the serine 2653 of the HTT peptide. CDK1 is highlighted by blue rectangles and CDK5/p25 by red rectangles. RBC, red blood cell substrate. Dotted lines indicate the 5% cut-off of enzyme activity used for the kinase screening.</p>
Full article ">Figure 2
<p>HTT peptides as potential as targets for CDKs. (<b>A</b>) The amino acid sequences surrounding serine of 1181, 1201, and 2653 of HTT from different species. S/T-P-x-K/R is a consensus site for both CDK1 and CDK5, which means that the amino acid of S or T followed by P-x-K/R could be phosphorylated by CDK1 and CDK5. (S = serine; T = threonine; P = proline; K = lysine; R = arginine; x is any amino acid). It is well conserved in vertebrates for S1181 and S1201 of the HTT sequence, but not S2653. (<b>B</b>) CDKs target serine 1181 of the HTT peptide. (<b>C</b>) CDKs target serine 1201 of HTT peptide. CDK1 is highlighted by blue rectangles and CDK5/p25 by red rectangles. RBC, red blood cell substrate. Dotted lines indicate the 1% cut-off of enzyme activity used for the kinase screening.</p>
Full article ">Figure 3
<p>The effect of knocking down CDK5 on the phosphorylation of S1181 and S1201 of mHTT. HEK293 cells were co-transfected with a plasmid expressing full-length mutant huntingtin with 82Q (FL-82Q) and siRNA targeting human CDK5 for 24 h. The expression level of CDK5, phosphorylated S1181-HTT (p-S1181), and phosphorylated S1201-HTT (p-S1201) were detected by western blot. (<b>A</b>) The representative western blot for CDK5. (<b>B</b>) The quantification of CDK5 expression. (<b>C</b>) The representative western blot for p-S1181-HTT. (<b>D</b>) The quantification of p-S1181-HTT expression. (<b>E</b>) The representative western blot for p-S1201-HTT. (<b>F</b>) The quantification of p-S1201-HTT expression. The experiment was repeated by at least two different analysts. Each experiment had <span class="html-italic">n</span> = 3. One representative experiment is presented. 2166 is the anti-HTT antibody (MAB2166, Millipore). RNAi = pooled siRNA targeting human CDK5. A two-tailed Student <span class="html-italic">t</span>-test was used. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>The effect of CDK5 overexpression on S1181 of mHTT. HEK293 cells were co-transfected with HTT plasmid FL-82Q and a plasmid expressing human CDK5 for 24 h. The expression levels of CDK5, p-S1181, and p-S1201 were detected by western blot. (<b>A</b>) The representative western blot for CDK5, p-S1181, 2166, and MW1. (<b>B</b>) The quantification of CDK5 expression. (<b>C</b>,<b>D</b>) The quantification of p-S1181 expression. (<b>E</b>) The quantification of total huntingtin (MAB2166) expression. (<b>F</b>) The quantification of mutant huntingtin (MW1) expression. The experiment was repeated by at least two different analysts. Each experiment had n = 3. One representative experiment is presented. 2166 is the anti-HTT antibody (MAB2166). MW1 is the anti-HTT antibody that binds to mutant huntingtin (clone MW1). RNAi = siRNA targeting human CDK5. A two-tailed Student <span class="html-italic">t</span>-test was used. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>CDK5 level altered cell toxicity induced by mHTT. The effect of CDK5 overexpression or knocking down was evaluated in mouse striatal cells with different poly Q (SThdh <sup>Q7/Q7</sup> or SThdh <sup>Q111/Q111</sup>). Cells were transfected with either a plasmid expressing human CDK5 or pooled siRNA targeting mouse CDK5 for 24 h. Cell death was measured with CytoTox-Glo cytotoxicity assay kit (Promega). The experiment was repeated by at least two different analysts. Each experiment had n = 3. One representative experiment is presented. Two-way ANOVA with Tukey’s multiple comparation was used for analysis. * <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.</p>
Full article ">Figure 6
<p>Phospho-null modification of S1181 and S1201 of mHTT altered the mHTT-induced cell toxicity. S1181 and S1201 in the plasmid of FL-82Q were artificially modified to alanine (A). (<b>A</b>) Modified plasmids were transiently transfected into HEK293 cells for 72 h, and cell toxicity was measured by caspase3/7 activity. (<b>B</b>) The modified plasmids were transient transfected in primary cortical neurons, and cell death was analyzed by nuclei condensation assay. The experiment was repeated by at least two different analysts. Each experiment had n = 3. One representative experiment is presented. One-way ANOVA with Dunnett’s multiple comparation was used for analysis. * <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.</p>
Full article ">Figure 7
<p>Roscovitine protected cells from mHTT-induced toxicity. Roscovitine was used in different HD cell models to evaluate its effect on mHTT-induced toxicity. (<b>A</b>). The effect of roscovitine on mHTT-induced toxicity in SThdh cells expressing Q111 cells. SThdh <sup>Q111/Q111</sup> cells were treated with roscovitine for 24 h under serum withdrawal conditions, and cell toxicity was measured using a CytoTox kit. (<b>B</b>). The effect of roscovitine on mHTT-induced toxicity in primary cortical neuronal HD cell model. Primary cultured cortical neurons were transiently transfected with plasmids expressing either normal Q (23Q) or poly Q (82Q) for 4 h and then treated with or without roscovitine for 48 h. A nuclei condensation assay was used to evaluate cell death. The experiment was repeated by at least two different analysts. Each experiment had <span class="html-italic">n</span> = 3. One representative experiment is presented. W/O = serum withdrawal. One-way ANOVA with Dunnett’s multiple comparation was used for analysis. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 8
<p>Roscovitine reduced the phosphorylation of S1181 and S1201 of mHTT. Roscovitine reduced p-S1181-HTT and p-S1201-HTT in vitro by inhibiting CDK5 but not CDK1. (<b>A</b>) The representative western blot for p-S1181 in HEK293 cells transfected with a plasmid expressing FL-82Q and treated with or without roscovitine for 48 h. (<b>B</b>) The representative western blot for p-S1201 in HEK293 cells transfected with a plasmid expressing FL-82Q and treated with or without roscovitine for 48 h. (<b>C</b>) The representative western blot for CDK5 in HEK293 cells transfected with a plasmid expressing FL-82Q and treated with or without roscovitine for 48 h. (<b>D</b>) The representative western blot for CDK1 in HEK293 cells transfected with a plasmid expressing FL-82Q and treated with or without roscovitine for 48 h. (<b>E</b>) Quantification of p-S1181 in A. (<b>F</b>) Quantification of p-S1201 in B. (<b>G</b>) Quantification of CDK5 in C. (<b>H</b>) Quantification of CDK1 in D. The experiment was repeated by at least two different analysts. Each experiment had n = 3. One representative experiment is presented. Ros~ = Roscovitine. A two-tailed Student <span class="html-italic">t</span>-test was used. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 9
<p>Roscovitine penetrated the mouse brain and inhibited CDK5 activity in the zQ175 HD mouse brain. Variant doses of roscovitine were injected into 4-month-old mice through intraperitoneal injection (IP). Plasma and brain samples were collected at different time points after injection. The concentration of roscovitine was measured by LCMS/MS. (<b>A</b>) Brain concentration of roscovitine at different time points after acute IP injection with variant doses. Each group has 4–5 mice. (<b>B</b>) The CDK5 activity in the brain of mice injected with roscovitine for three weeks. Two doses of roscovitine were injected into 4-month-old zQ175HD mice by IP daily for three weeks. Twenty-four hours after the last injection, the mouse brain samples were collected, and the CDK5 activity was measured using a commercial kit from Promega. Animal number is indicated in the bar graph for each group. R-25 = 25 mg/kg of roscovitine. R-50 = 50 mg/kg of roscovitine. One-way ANOVA with Fish’s LSD was used for analysis. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 10
<p>Tolerability of mice injected with roscovitine by IP administration. (<b>A</b>) The body weight loss of zQ175 HD mice injected with roscovitine. (<b>B</b>) The stride length (by CatWalk) in zQ175 HD mice before and after being injected with roscovitine. Roscovitine was injected by IP daily for three weeks. Animal number for each group is indicated in the bar graphs. V = Vehicle, R = roscovitine. R-25 = 25 mg/kg of roscovitine. R-50 = 50 mg/kg of roscovitine. One-way ANOVA with Fish’s LSD was used for analysis.</p>
Full article ">Figure 11
<p>Molecular neurobiological measurement in the brain of zQ175 HD mice injected with roscovitine by IP. Brain samples were collected as previously described. The cortex was dissected, and lysates were made for western blot analysis. (<b>A</b>) The expression of a pan-neuronal marker, NeuN. (<b>B</b>) The expression of astrocyte marker GFAP. (<b>C</b>) The expression of microglial marker IBA1. (<b>D</b>) The expression of mHTT marker MW1. (<b>E</b>) The quantification of NeuN. (<b>F</b>) The quantification of GFAP. (<b>G</b>) The quantification of IBA1. (<b>H</b>) The quantification of MW1. Animal number is indicated in the bar graph for each group. Veh~ = Vehicle, R25 = 25 mg/kg of roscovitine. R50 = 50 mg/kg of roscovitine. One-way ANOVA with Fish’s LSD was used for analysis.</p>
Full article ">
25 pages, 1292 KiB  
Review
Designed Cell-Penetrating Peptide Constructs for Inhibition of Pathogenic Protein Self-Assembly
by Mona Kalmouni, Yujeong Oh, Wael Alata and Mazin Magzoub
Pharmaceutics 2024, 16(11), 1443; https://doi.org/10.3390/pharmaceutics16111443 - 12 Nov 2024
Viewed by 1022
Abstract
Peptides possess a number of pharmacologically desirable properties, including greater chemical diversity than other biomolecule classes and the ability to selectively bind to specific targets with high potency, as well as biocompatibility, biodegradability, and ease and low cost of production. Consequently, there has [...] Read more.
Peptides possess a number of pharmacologically desirable properties, including greater chemical diversity than other biomolecule classes and the ability to selectively bind to specific targets with high potency, as well as biocompatibility, biodegradability, and ease and low cost of production. Consequently, there has been considerable interest in developing peptide-based therapeutics, including amyloid inhibitors. However, a major hindrance to the successful therapeutic application of peptides is their poor delivery to target tissues, cells or subcellular organelles. To overcome these issues, recent efforts have focused on engineering cell-penetrating peptide (CPP) antagonists of amyloidogenesis, which combine the attractive intrinsic properties of peptides with potent therapeutic effects (i.e., inhibition of amyloid formation and the associated cytotoxicity) and highly efficient delivery (to target tissue, cells, and organelles). This review highlights some promising CPP constructs designed to target amyloid aggregation associated with a diverse range of disorders, including Alzheimer’s disease, transmissible spongiform encephalopathies (or prion diseases), Parkinson’s disease, and cancer. Full article
(This article belongs to the Special Issue State of the Art of Membrane Active Peptides, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Designed cell-penetrating peptide (CPP) constructs inhibit amyloid aggregation and the associated cytotoxicity. Amyloid inhibitor CPP constructs are typically composed of distinct segments that contribute to the therapeutic effects (i.e., inhibition of oligomerization, fiber formation, and the associated cytotoxicity) and/or delivery properties (to target tissue, cells, and subcellular organelles). An example is NCAM1-PrP, which is composed of a hydrophobic signal peptide from the neural cell adhesion molecule-1 (NCAM1<sub>1–19</sub>: MLRTKDLIWTLFFLGTAVS) followed by a cationic nuclear localization signal (NLS)-like hexapeptide sequence from the prion protein (PrP<sub>23–28</sub>: KKRPKP) [<a href="#B149-pharmaceutics-16-01443" class="html-bibr">149</a>]. NCAM1-PrP was shown to effectively inhibit conversion of normal PrP<sup>C</sup> into its disease-associated scrapie isoform of the protein (PrP<sup>Sc</sup>) [<a href="#B149-pharmaceutics-16-01443" class="html-bibr">149</a>], and to antagonize Aβ oligomerization, fiber formation, and the associated neurotoxicity [<a href="#B150-pharmaceutics-16-01443" class="html-bibr">150</a>]. The inhibition of pathogenic protein self-assembly is attributed to the NLS-like hexapeptide, but this sequence alone is poorly cell-permeable and requires the hydrophobic NCAM1 signal peptide to acquire its CPP properties [<a href="#B165-pharmaceutics-16-01443" class="html-bibr">165</a>]. Created in <a href="http://BioRender" target="_blank">BioRender</a>. Oh, Y. (accessed on 7 October 2024) <a href="http://BioRender.com/k09j761" target="_blank">BioRender.com/k09j761</a>.</p>
Full article ">Figure 2
<p>Amyloid inhibitors potently abrogate cancer-associated mutant p53 aggregation and restore tumor suppressor function. Under physiological conditions, p53 protein exists as a homotetramer, with each monomer composed of discrete domains for DNA binding, tetramerization, and transcriptional activation [<a href="#B212-pharmaceutics-16-01443" class="html-bibr">212</a>]. Approximately 90% of the cancer-associated p53 mutations occur within the thermodynamically unstable DNA-binding domain (DBD) [<a href="#B212-pharmaceutics-16-01443" class="html-bibr">212</a>,<a href="#B215-pharmaceutics-16-01443" class="html-bibr">215</a>], many of which decrease the domain’s stability further and prompt its unfolding and self-assembly into amyloid-like aggregates within inactive cytosolic inclusions [<a href="#B164-pharmaceutics-16-01443" class="html-bibr">164</a>,<a href="#B214-pharmaceutics-16-01443" class="html-bibr">214</a>,<a href="#B217-pharmaceutics-16-01443" class="html-bibr">217</a>]. Similar to the CPP construct ReACp53 [<a href="#B164-pharmaceutics-16-01443" class="html-bibr">164</a>], the protein mimetic ADH-6 (a cationic tripyridylamide) efficiently enters cancer cells, where it targets aggregation-prone p53 mutants and potently abrogates their self-assembly, which shifts the folding equilibrium towards the soluble state, leading to dissociation of the inactive cytosolic inclusions and accumulation of functional protein in the nucleus [<a href="#B229-pharmaceutics-16-01443" class="html-bibr">229</a>].</p>
Full article ">
21 pages, 2912 KiB  
Article
Role of Peptide Associations in Enhancing the Antimicrobial Activity of Adepantins: Comparative Molecular Dynamics Simulations and Design Assessments
by Matko Maleš, Davor Juretić and Larisa Zoranić
Int. J. Mol. Sci. 2024, 25(22), 12009; https://doi.org/10.3390/ijms252212009 - 8 Nov 2024
Viewed by 808
Abstract
Adepantins are peptides designed to optimize antimicrobial biological activity through the choice of specific amino acid residues, resulting in helical and amphipathic structures. This paper focuses on revealing the atomistic details of the mechanism of action of Adepantins and aligning design concepts with [...] Read more.
Adepantins are peptides designed to optimize antimicrobial biological activity through the choice of specific amino acid residues, resulting in helical and amphipathic structures. This paper focuses on revealing the atomistic details of the mechanism of action of Adepantins and aligning design concepts with peptide behavior through simulation results. Notably, Adepantin-1a exhibits a broad spectrum of activity against both Gram-positive and Gram-negative bacteria, while Adepantin-1 has a narrow spectrum of activity against Gram-negative bacteria. The simulation results showed that one of the main differences is the extent of aggregation. Both peptides exhibit a strong tendency to cluster due to the amphipathicity embedded during design process. However, the more potent Adepantin-1a forms smaller aggregates than Adepantin-1, confirming the idea that the optimal aggregations, not the strongest aggregations, favor activity. Additionally, we show that incorporation of the cell penetration region affects the mechanisms of action of Adepantin-1a and promotes stronger binding to anionic and neutral membranes. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structures of Adepantin-1 (<b>on the left</b>), Adepantin-1a <b>(in the middle</b>), and CPP-Adepantin-1a (<b>on the right</b>) as predicted by the C-QUARK structure predictor (<a href="https://zhanggroup.org/C-QUARK/" target="_blank">https://zhanggroup.org/C-QUARK/</a>, accessed on 9 February 2024). The side and top views show the peptides represented by ribbon and point models. Hydrophobic residues are shown in magenta, positively charged residues in yellow, negatively charged residues in cyan, histidine in purple, serine in orange, and glycine in gray. The bottom row presents the 2D helical wheel projections with the same residue colors (<a href="https://heliquest.ipmc.cnrs.fr/cgi-bin/ComputParams.py" target="_blank">https://heliquest.ipmc.cnrs.fr/cgi-bin/ComputParams.py</a>, accessed on 10 February 2024). More information from the Heliquest server is shown in <a href="#app1-ijms-25-12009" class="html-app">Supplementary Figure S1</a>.</p>
Full article ">Figure 2
<p>Characteristic states during peptide binding to the anionic membrane and corresponding vectors of the 3D hydrophobic moment (black) and electrostatic dipole moment vectors (blue) for Ad-1 (<b>left</b>), Ad-1a (<b>middle</b>), and CPP-Ad-1a (<b>right</b>). The first row (<b>a</b>) shows the initial electrostatic binding. The second row (<b>b</b>) shows the intermediate state for reorienting hydrophobic and hydrophilic parts. This reorientation is limited in CPP-Ad-1a due to the anchoring of charged CPP regions. The third row (<b>c</b>) shows final states, with hydrophobic parts facing the membrane in Adepantins and more complex behavior in CPP-Ad-1a. Peptides are shown in ribbon and stick–ball forms, with hydrophilic residues in yellow and hydrophobic residues in magenta. Only the upper membrane leaflet is shown, with phosphorus atoms (P) in green (POPE) and orange (POPG). Other lipid atoms and water are omitted for clarity. Vector sizes are detailed in <a href="#app1-ijms-25-12009" class="html-app">Tables S1 and S2 in the Supplementary Materials</a>.</p>
Full article ">Figure 3
<p>Distance between the center of mass and the membrane center for charged and hydrophobic residues for Adepantin-1 (<b>left</b>), Adepantin-1a (<b>middle</b>), and CPP-Adepantin-1a (<b>right</b>) in AA-1 simulations. More results are presented in <a href="#app1-ijms-25-12009" class="html-app">Figure S2 in the Supplementary Materials</a>.</p>
Full article ">Figure 4
<p>Top-view configurations and clustering of twelve peptides interacting with an anionic membrane. The simulation snapshots are taken at t = 1 μs for AA (<b>top row</b>) and t = 25 μs for CG simulations (<b>bottom row</b>). The coloring scheme for the residues is as follows: charged and polar residues are highlighted in yellow, while hydrophobic residues are shown in magenta. Small spheres represent phosphorus (P) atoms within the lipid molecules, where green spheres correspond to POPE lipids and orange spheres to POPG lipids. To enhance clarity, water molecules and other lipid atoms have been omitted from the images.</p>
Full article ">Figure 5
<p>Results of cluster analysis for the AA-12 simulations of Ad-1 (<b>left</b>), Ad-1a (<b>middle</b>), and CPP-Ad-1a (<b>right</b>) peptides. The (<b>top row</b>) presents the time dependence of the number of clusters and the number of peptides in the largest cluster throughout the simulation. The (<b>bottom row</b>) displays the cluster size distributions.</p>
Full article ">Figure 6
<p>Results of cluster analysis for the CG-12 simulations of Ad-1 (<b>left</b>), Ad-1a (<b>middle</b>), and CPP-Ad-1a (<b>right</b>) peptides. The <b>top row</b> presents the time dependence of the number of clusters and the number of peptides in the largest cluster throughout the simulation. The <b>bottom row</b> displays the cluster size distributions.</p>
Full article ">Figure 7
<p>Results of the residue-pair distance analysis for the simulations of Adepantins AA-12. Parts (<b>a</b>,<b>b</b>) show the distance matrices, consisting of the smallest distances between residue pairs of all twelve peptides (a total of 23 × 12 = 276 residues), for the case1 simulations of Ad-1 (<b>a</b>) and Ad-1a (<b>b</b>). Part (<b>c</b>) presents the average number of contacts per residue for the last 100 ns of simulation time in each simulation case. Small motifs in Ad-1 sequence are marked in brown (GXXXG), while the small motifs in Ad-1a sequence are in red/green (GXXXA) and blue (AXXXA). Other results are presented in <a href="#app1-ijms-25-12009" class="html-app">Supplementary Figures S7 and S8</a>.</p>
Full article ">
28 pages, 3339 KiB  
Review
Enhancing Tumor Targeted Therapy: The Role of iRGD Peptide in Advanced Drug Delivery Systems
by Dragana Nikitovic, Ekaterina Kukovyakina, Aikaterini Berdiaki, Alexandros Tzanakakis, Anna Luss, Elizaveta Vlaskina, Anne Yagolovich, Aristides Tsatsakis and Andrey Kuskov
Cancers 2024, 16(22), 3768; https://doi.org/10.3390/cancers16223768 - 8 Nov 2024
Viewed by 1860
Abstract
Chemotherapy remains the primary therapeutic approach in treating cancer. The tumor microenvironment (TME) is the complex network surrounding tumor cells, comprising various cell types, such as immune cells, fibroblasts, and endothelial cells, as well as ECM components, blood vessels, and signaling molecules. The [...] Read more.
Chemotherapy remains the primary therapeutic approach in treating cancer. The tumor microenvironment (TME) is the complex network surrounding tumor cells, comprising various cell types, such as immune cells, fibroblasts, and endothelial cells, as well as ECM components, blood vessels, and signaling molecules. The often stiff and dense network of the TME interacts dynamically with tumor cells, influencing cancer growth, immune response, metastasis, and resistance to therapy. The effectiveness of the treatment of solid tumors is frequently reduced due to the poor penetration of the drug, which leads to attaining concentrations below the therapeutic levels at the site. Cell-penetrating peptides (CPPs) present a promising approach that improves the internalization of therapeutic agents. CPPs, which are short amino acid sequences, exhibit a high ability to pass cell membranes, enabling them to deliver drugs efficiently with minimal toxicity. Specifically, the iRGD peptide, a member of CPPs, is notable for its capacity to deeply penetrate tumor tissues by binding simultaneously integrins ανβ3/ανβ5 and neuropilin receptors. Indeed, ανβ3/ανβ5 integrins are characteristically expressed by tumor cells, which allows the iRGD peptide to home onto tumor cells. Notably, the respective dual-receptor targeting mechanism considerably increases the permeability of blood vessels in tumors, enabling an efficient delivery of co-administered drugs or nanoparticles into the tumor mass. Therefore, the iRGD peptide facilitates deeper drug penetration and improves the efficacy of co-administered therapies. Distinctively, we will focus on the iRGD mechanism of action, drug delivery systems and their application, and deliberate future perspectives in developing iRGD-conjugated therapeutics. In summary, this review discusses the potential of iRGD in overcoming barriers to drug delivery in cancer to maximize treatment efficiency while minimizing side effects. Full article
(This article belongs to the Special Issue Tumor Microenvironment: Intercellular Communication)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of RGD and iRGD peptides.</p>
Full article ">Figure 2
<p>Tumor microenvironment. The TME consists of tumor and stromal cells, including lymphocytes, cancer-associated fibroblasts, and immune suppressor cells, surrounded by a dense, stiff, intensely remodeled ECM. It is characterized by hypoxia, acidic conditions, increased interstitial pressure, and intense angiogenesis, which results in poorly formed blood vessels. These physical, chemical, and biological features present considerable drug penetration barriers. Cancer cells exhibit discrete integrin expression compared to healthy cells. Created in BioRender. Nikitovic, D. (2024), BioRender.com/l29i302 (accessed on 30 October 2024).</p>
Full article ">Figure 3
<p>The roles of ανβ3/ανβ5 in the facilitation of carcinogenesis. Created in BioRender. Nikitovic, D. (2024), BioRender.com/c83e444 (accessed on 2 October 2024).</p>
Full article ">Figure 4
<p>The iRGD mechanism of cargo delivery. The iRGD peptide carrying drug cargo binds to integrin β3/β5 through the RGD motif. This facilitates the cleavage of the CDGR sequence and enhances the binding of the remaining CendR motif carrying cargo to NRP1. CendR/NRP-1 binding improves cargo uptake by endothelial and tumor cells expressing β3/β5 integrin. Created in BioRender. Nikitovic, D. (2024), BioRender.com/y62t355 (accessed on 2 October 2024).</p>
Full article ">Figure 5
<p>Mechanisms of iRGD conjugation.</p>
Full article ">Figure 6
<p>Attachment of the iRGD peptide to a pegylated lipid.</p>
Full article ">Figure 7
<p>An outline of a workflow of AI utilization in peptide discovery and improvement for tumor targeting. The process involves gathering data on existing peptides, including their sequences, structures, interactions with tumor cells, and experimental results. The data are then cleaned, standardized, and preprocessed. Features such as amino acid composition and physicochemical properties are extracted. AI models are trained to predict effective peptide sequences and validated using known peptides. Visualization of predictions helps compare peptide efficacy, and interpretation guides decisions on which peptides to synthesize and test. The feedback loop integrates experimental results to refine the model, resulting in an AI tool capable of generating or improving tumor-targeting peptides ready for research or clinical use.</p>
Full article ">
16 pages, 1041 KiB  
Review
Research on the Mechanisms of Phytohormone Signaling in Regulating Root Development
by Yuru Ma, Ying Zhang, Jiahui Xu, Jiahong Qi, Xigang Liu, Lin Guo and Hao Zhang
Plants 2024, 13(21), 3051; https://doi.org/10.3390/plants13213051 - 31 Oct 2024
Viewed by 997
Abstract
Phytohormones are organic compounds produced in trace amounts within plants that regulate their physiological processes. Their physiological effects are highly complex and diverse. They influence processes ranging from cell division, elongation, and differentiation to plant germination and rooting. Therefore, phytohormones play a crucial [...] Read more.
Phytohormones are organic compounds produced in trace amounts within plants that regulate their physiological processes. Their physiological effects are highly complex and diverse. They influence processes ranging from cell division, elongation, and differentiation to plant germination and rooting. Therefore, phytohormones play a crucial regulatory role in plant growth and development. Recently, various studies have highlighted the role of PHs, such as auxin, cytokinin (CK), and abscisic acid (ABA), and newer classes of PHs, such as brassinosteroid (BR) and peptide hormone, in the plant responses toward environmental stresses. These hormones not only have distinct roles at different stages of plant growth but also interact to promote or inhibit each other, thus effectively regulating plant development. Roots are the primary organs for water and mineral absorption in plants. During seed germination, the radicle breaks through the seed coat and grows downward to form the primary root. This occurs because the root needs to quickly penetrate the soil to absorb water and nutrients, providing essential support for the plant’s subsequent growth. Root development is a highly complex and precisely regulated process influenced by various signals. Changes in root architecture can affect the plant’s ability to absorb nutrients and water, which in turn impacts crop yield. Thus, studying the regulation of root development is of great significance. Numerous studies have reported on the role of phytohormones, particularly auxins, in root regulation. This paper reviews recent studies on the regulation of root development by various phytohormones, both individually and in combination, providing a reference for researchers in this field and offering perspectives on future research directions for improving crop yields. Full article
Show Figures

Figure 1

Figure 1
<p>Regulatory network of the auxin involved in regulating root development. Light, energy, and auxin coordinate the regulation of root development.</p>
Full article ">Figure 2
<p>Regulatory network of the auxin involved in regulating lateral root development.</p>
Full article ">Figure 3
<p>Regulatory network of the CK involved in regulating root function and maintenance.</p>
Full article ">Figure 4
<p>Regulatory network of the BR crosstalk involved in regulating root growth.</p>
Full article ">
26 pages, 1754 KiB  
Review
Unlocking Genome Editing: Advances and Obstacles in CRISPR/Cas Delivery Technologies
by Bibifatima Kaupbayeva, Andrey Tsoy, Yuliya Safarova (Yantsen), Ainetta Nurmagambetova, Hironobu Murata, Krzysztof Matyjaszewski and Sholpan Askarova
J. Funct. Biomater. 2024, 15(11), 324; https://doi.org/10.3390/jfb15110324 - 31 Oct 2024
Viewed by 3355
Abstract
CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats associated with protein 9) was first identified as a component of the bacterial adaptive immune system and subsequently engineered into a genome-editing tool. The key breakthrough in this field came with the realization that CRISPR/Cas9 could [...] Read more.
CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats associated with protein 9) was first identified as a component of the bacterial adaptive immune system and subsequently engineered into a genome-editing tool. The key breakthrough in this field came with the realization that CRISPR/Cas9 could be used in mammalian cells to enable transformative genetic editing. This technology has since become a vital tool for various genetic manipulations, including gene knockouts, knock-in point mutations, and gene regulation at both transcriptional and post-transcriptional levels. CRISPR/Cas9 holds great potential in human medicine, particularly for curing genetic disorders. However, despite significant innovation and advancement in genome editing, the technology still possesses critical limitations, such as off-target effects, immunogenicity issues, ethical considerations, regulatory hurdles, and the need for efficient delivery methods. To overcome these obstacles, efforts have focused on creating more accurate and reliable Cas9 nucleases and exploring innovative delivery methods. Recently, functional biomaterials and synthetic carriers have shown great potential as effective delivery vehicles for CRISPR/Cas9 components. In this review, we attempt to provide a comprehensive survey of the existing CRISPR-Cas9 delivery strategies, including viral delivery, biomaterials-based delivery, synthetic carriers, and physical delivery techniques. We underscore the urgent need for effective delivery systems to fully unlock the power of CRISPR/Cas9 technology and realize a seamless transition from benchtop research to clinical applications. Full article
(This article belongs to the Special Issue Medical Application of Functional Biomaterials (2nd Edition))
Show Figures

Figure 1

Figure 1
<p>Schematic representation of CRISPR/Cas9-mediated gene editing. DSB—double-strand break, NHEJ—nonhomologous end joining, HDR—homology-directed repair, ssDNA—single-stranded DNA, dsDNA—double-stranded DNA.</p>
Full article ">Figure 2
<p>Biomaterials-based carriers used for the delivery of CRISPR/Cas9 systems.</p>
Full article ">Figure 3
<p>Schematic on the synthesis of LNP for CRISPR/Cas9 delivery system.</p>
Full article ">Figure 4
<p>“Grafting from” Cas9. (<b>A</b>) ATRP-synthesis of Cas9-polymer conjugates. (<b>B</b>) Delivery of Cas9-polymer conjugates into cells.</p>
Full article ">
21 pages, 3811 KiB  
Review
The Influence of Cholesterol on Membrane Targeted Bioactive Peptides: Modulating Peptide Activity Through Changes in Bilayer Biophysical Properties
by Juan M. Giraldo-Lorza, Chad Leidy and Marcela Manrique-Moreno
Membranes 2024, 14(10), 220; https://doi.org/10.3390/membranes14100220 - 17 Oct 2024
Cited by 1 | Viewed by 1547
Abstract
Cholesterol is a biological molecule that is essential for cellular life. It has unique features in terms of molecular structure and function, and plays an important role in determining the structure and properties of cell membranes. One of the most recognized functions of [...] Read more.
Cholesterol is a biological molecule that is essential for cellular life. It has unique features in terms of molecular structure and function, and plays an important role in determining the structure and properties of cell membranes. One of the most recognized functions of cholesterol is its ability to increase the level of lipid packing and rigidity of biological membranes while maintaining high levels of lateral mobility of the bulk lipids, which is necessary to sustain biochemical signaling events. There is increased interest in designing bioactive peptides that can act as effective antimicrobial agents without causing harm to human cells. For this reason, it becomes relevant to understand how cholesterol can affect the interaction between bioactive peptides and lipid membranes, in particular by modulating the peptides’ ability to penetrate and disrupt the membranes through these changes in membrane rigidity. Here we discuss cholesterol and its role in modulating lipid bilayer properties and discuss recent evidence showing how cholesterol modulates bioactive peptides to different degrees. Full article
Show Figures

Figure 1

Figure 1
<p>Secondary structures of representative BAPs using PyMOL 3.0. In brackets are the PDB ID and the origin of the peptides. The colors represent the secondary structures: (<b>a</b>) Indolicidin (PDB ID 8IS3, <span class="html-italic">Bos taurus</span>), (<b>b</b>) Magainin 2 (PDB ID 2MAG, <span class="html-italic">Xenopus laevis</span>), (<b>c</b>) Human β-Defensin-4 (PDB ID 5KI9, <span class="html-italic">Homo sapiens</span>), and (<b>d</b>) Human β-Defensin-2 (PDB ID 1FD4, <span class="html-italic">Homo sapiens</span>).</p>
Full article ">Figure 2
<p>Representation of eukaryotic cell plasma membrane. The phospholipid bilayer contains all molecules, including phospholipids, proteins, and cholesterol. Phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS), and sphingomyelin (SM). The irregular representation of the lipid acyl chains of lipids denotes the fluid nature of the bilayer.</p>
Full article ">Figure 3
<p>Chemical representation of the head groups and acyl chains of the most abundant phospholipids of eukaryotic cell membranes. Phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS), sphingomyelin (SM), and phosphatidylinositol (PI). The acyl chains range from fully saturated to multiple unsaturated.</p>
Full article ">Figure 4
<p>The lipid composition of different organelles throughout the eukaryotic cell. The lipid data in the graphs are presented as a percentage of total phospholipids (PL) in mammals. The cholesterol content is presented as the molar ratio of cholesterol (CHO) with respect to the PL [<a href="#B2-membranes-14-00220" class="html-bibr">2</a>,<a href="#B3-membranes-14-00220" class="html-bibr">3</a>,<a href="#B61-membranes-14-00220" class="html-bibr">61</a>]. * Data was reported in CHO/PL ratio.</p>
Full article ">Figure 5
<p>Summary of publications that quantified the phospholipid distribution in mammalian cells. Others are fibroblasts and cancer cells. The colors correspond to PC (<span class="html-fig-inline" id="membranes-14-00220-i001"><img alt="Membranes 14 00220 i001" src="/membranes/membranes-14-00220/article_deploy/html/images/membranes-14-00220-i001.png"/></span>), SM (<span class="html-fig-inline" id="membranes-14-00220-i002"><img alt="Membranes 14 00220 i002" src="/membranes/membranes-14-00220/article_deploy/html/images/membranes-14-00220-i002.png"/></span>), PE (<span class="html-fig-inline" id="membranes-14-00220-i003"><img alt="Membranes 14 00220 i003" src="/membranes/membranes-14-00220/article_deploy/html/images/membranes-14-00220-i003.png"/></span>), and PS (<span class="html-fig-inline" id="membranes-14-00220-i004"><img alt="Membranes 14 00220 i004" src="/membranes/membranes-14-00220/article_deploy/html/images/membranes-14-00220-i004.png"/></span>). Data are presented in ratios based on PC abundance.</p>
Full article ">Figure 6
<p>(<b>a</b>) Chemical structure of cholesterol and (<b>b</b>) disposition of cholesterol into the hydrophobic region of the cell membrane.</p>
Full article ">
Back to TopTop