[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (47)

Search Parameters:
Keywords = helper lipid

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 273 KiB  
Article
Metabolic and Immune Parameters in Pregnant Women with Impaired Glucose Metabolism—A Pilot Study
by Jelena Omazić, Andrijana Muller, Blaž Dumančić, Mirta Kadivnik, Jasna Aladrović, Lana Pađen, Kristina Kralik, Nikolina Brkić, Blaženka Dobrošević, Barbara Vuković and Jasenka Wagner
Metabolites 2024, 14(10), 551; https://doi.org/10.3390/metabo14100551 - 16 Oct 2024
Viewed by 286
Abstract
Gestational diabetes mellitus (GDM) is a public health problem with increasing prevalence. Analyses of metabolic and immune profiles have great potential for discovering new markers and mechanisms related to the development of GDM. We monitored 61 pregnant women during the first and third [...] Read more.
Gestational diabetes mellitus (GDM) is a public health problem with increasing prevalence. Analyses of metabolic and immune profiles have great potential for discovering new markers and mechanisms related to the development of GDM. We monitored 61 pregnant women during the first and third trimesters of pregnancy, including 13 pregnant women with GDM, 14 pregnant women with elevated glucose in the first trimester and 34 healthy pregnant women. A number of metabolic and immunological parameters were measured, including glucose, insulin, lipid status, fatty acids, lymphocyte profile, adiponectin, IL-6, IL-10 and TNF-a. A higher number of T-helper lymphocytes and a higher ratio of helper/cytotoxic lymphocytes was found in the control group in the first trimester of pregnancy. Pregnant women whose glucose threshold values were measured in the first trimester, but who did not develop GDM, showed a higher percentage of neutrophils and a lower percentage of lymphocytes in the third trimester. Differences in polyunsaturated fatty acids levels were observed between healthy pregnant women and those with glucose metabolism disorders in the first trimester of pregnancy. The results of this pilot study demonstrate that there are differences in the profiles of T lymphocytes, NK cells and polyunsaturated fatty acids between the examined groups of pregnant women, which can serve as a direction for future research. Full article
(This article belongs to the Special Issue Glucose Metabolism in Pregnancy)
19 pages, 3026 KiB  
Article
Stable Polymer-Lipid Hybrid Nanoparticles Based on mcl-Polyhydroxyalkanoate and Cationic Liposomes for mRNA Delivery
by Sergey M. Shishlyannikov, Ilya N. Zubkov, Vera V. Vysochinskaya, Nina V. Gavrilova, Olga A. Dobrovolskaya, Ekaterina A. Elpaeva, Mikhail A. Maslov and Andrey Vasin
Pharmaceutics 2024, 16(10), 1305; https://doi.org/10.3390/pharmaceutics16101305 - 7 Oct 2024
Viewed by 688
Abstract
Background/Objectives: The development of polymer–lipid hybrid nanoparticles (PLNs) is a promising area of research, as it can help increase the stability of cationic lipid carriers. Hybrid PLNs are core–shell nanoparticle structures that combine the advantages of both polymer nanoparticles and liposomes, especially in [...] Read more.
Background/Objectives: The development of polymer–lipid hybrid nanoparticles (PLNs) is a promising area of research, as it can help increase the stability of cationic lipid carriers. Hybrid PLNs are core–shell nanoparticle structures that combine the advantages of both polymer nanoparticles and liposomes, especially in terms of their physical stability and biocompatibility. Natural polymers such as polyhydroxyalkanoate (PHA) can be used as a matrix for the PLNs’ preparation. Methods: In this study, we first obtained stable cationic hybrid PLNs using a cationic liposome (CL) composed of a polycationic lipid 2X3 (1,26-bis(cholest-5-en-3β-yloxycarbonylamino)-7,11,16,20-tetraazahexacosane tetrahydrochloride), helper lipid DOPE (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine), and the hydrophobic polymer mcl-PHA, which was produced by the soil bacterium Pseudomonas helmantisensis P1. Results: The new polymer-lipid carriers effectively encapsulated and delivered model mRNA-eGFP (enhanced green fluorescent protein mRNA) to BHK-21 cells. We then evaluated the role of mcl-PHA in increasing the stability of cationic PLNs in ionic solutions using dynamic light scattering data, electrophoretic mobility, and transmission electron microscopy techniques. Conclusions: The results showed that increasing the concentration of PBS (phosphate buffered saline) led to a decrease in the stability of the CLs. At high concentrations of PBS, the CLs aggregate. In contrast, the presence of isotonic PBS did not result in the aggregation of PLNs, and the particles remained stable for 120 h when stored at +4 °C. The obtained results show that PLNs hold promise for further in vivo studies on nucleic acid delivery. Full article
(This article belongs to the Special Issue Polymer-Based Delivery System)
Show Figures

Figure 1

Figure 1
<p>Reactor for the synthesis of PLNs. Reaction tube and the sonicator probe are placed in a beaker with water. During the synthesis and under sonication, the organic solvent (<span class="html-italic">n</span>-hexane) is removed by evaporation in argon stream.</p>
Full article ">Figure 2
<p>Analysis of mRNA binding with PLNs by capillary electrophoresis. Control RNA—control mRNA samples without PLNs containing 50 ng of mRNA. PHA-2X3—complexes of mRNA with PLNs stabilized by 2X3; PHA-2X3-DOPE 1:1—complexes of mRNA with PLNs stabilized by a mixture of 2X3 and DOPE in a molar ratio of 1:1; PHA-2X3-DOPE 1:2—complexes of mRNA with PLNs stabilized with a mixture of 2X3 and DOPE in a molar ratio of 1:2. PHA-2X3-DOPE 1:3—complexes of mRNA with PLNs stabilized with a mixture of 2X3 and DOPE in a molar ratio of 1:3. PHA/lipids ratio in all the PLNs was 20:1 (wt.). Green line—signal of the fluorescent dye (used as an internal control for the electrophoresis). N/P — molar ratio between positively charged cationic liposomes and negatively charged delivered mRNA molecules.</p>
Full article ">Figure 3
<p>The efficiency of mRNA-eGFP delivery using CLs and PLNs in BHK-21 cells was measured using flow cytometry. The transfected cells were analyzed for the percentage of cells with detectable eGFP signals, and the MFI (mean fluorescence intensity) was recorded. Lipofectamine MessengerMAX (Lipofectamine MM) is a commercial transfection reagent and was used as a positive control for mRNA transfection. All the measurements were triplicated. The statistical analysis was performed using a two-way ANOVA: **—<span class="html-italic">p</span> &lt; 0.01; not significant—‘ns’.</p>
Full article ">Figure 4
<p>Dependences of the average particle diameter (d, nm), polydispersity (PDI), and ζ-potential (ζ, mV) on the N/P. Complexes of the PLNs (PHA-2X3-DOPE 1:3) and mRNA-eGFP were analyzed. PLNs (PHA-2X3-DOPE 1:3) were used as a control.</p>
Full article ">Figure 5
<p>(<b>a</b>) Efficiency of mRNA-Cy5 uptake with PLNs and CLs by BHK-21 cells determined by flow cytometry. Transfected cells—the percentage of fluorescent cells (Cy5 signal); MFI—mean fluorescence intensity. Statistical analysis was performed using one-way ANOVA: ***—<span class="html-italic">p</span> &lt; 0.001; **—<span class="html-italic">p</span> &lt; 0.01. (<b>b</b>) Fluorescence microscopy of cells transfected with mRNA-Cy5 mRNA complexes using PHA-2X3-DOPE 1:3, 2X3-DOPE 1:3, and Lipofectamine Messenger MAX (Lipofectamine MM); Scale bar 50 μm; cell nuclei–blue, eGFP—green, and mRNA–Cy5—red.</p>
Full article ">Figure 6
<p>Dependences of the average particle diameter (d, nm), polydispersity (PDI) and ζ-potential (ζ, mV) on the molar concentration of Na<sup>+</sup> in PBS. PLNs (PHA-2X3-DOPE 1:3) or CLs (2X3-DOPE 1:3) were mixed with PBS and incubated for either 2 h at 25 °C or 5 days at 4 °C.</p>
Full article ">Figure 7
<p>Transmission electron microscopy of PLNs (PHA-2X3-DOPE 1:3) and CLs (2X3-DOPE 1:3) at various Na<sup>+</sup> concentrations (0, 30, and 150 mM; incubation time 2 h). Scale bar: 150 nm (<b>A,B</b>), 100 nm (<b>C,E</b>), 200 nm (<b>D</b>), 500 nm (<b>F</b>).</p>
Full article ">
13 pages, 6837 KiB  
Article
Cationic Serine-Based Gemini Surfactant:Monoolein Aggregates as Viable and Efficacious Agents for DNA Complexation and Compaction: A Cytotoxicity and Physicochemical Assessment
by Isabel S. Oliveira, Sandra G. Silva, Andreia C. Gomes, M. Elisabete C. D. Real Oliveira, M. Luísa C. do Vale and Eduardo F. Marques
J. Funct. Biomater. 2024, 15(8), 224; https://doi.org/10.3390/jfb15080224 - 13 Aug 2024
Viewed by 947
Abstract
Cationic gemini surfactants have emerged as potential gene delivery agents as they can co-assemble with DNA due to a strong electrostatic association. Commonly, DNA complexation is enhanced by the inclusion of a helper lipid (HL), which also plays a key role in transfection [...] Read more.
Cationic gemini surfactants have emerged as potential gene delivery agents as they can co-assemble with DNA due to a strong electrostatic association. Commonly, DNA complexation is enhanced by the inclusion of a helper lipid (HL), which also plays a key role in transfection efficiency. The formation of lipoplexes, used as non-viral vectors for transfection, through electrostatic and hydrophobic interactions is affected by various physicochemical parameters, such as cationic surfactant:HL molar ratio, (+/−) charge ratio, and the morphological structure of the lipoplexes. Herein, we investigated the DNA complexation ability of mixtures of serine-based gemini surfactants, (nSer)2N5, and monoolein (MO) as a helper lipid. The micelle-forming serine surfactants contain long lipophilic chains (12 to 18 C atoms) and a five CH2 spacer, both linked to the nitrogen atoms of the serine residues by amine linkages. The (nSer)2N5:MO aggregates are non-cytotoxic up to 35–90 µM, depending on surfactant and surfactant/MO mixing ratio, and in general, higher MO content and longer surfactant chain length tend to promote higher cell viability. All systems efficaciously complex DNA, but the (18Ser)2N5:MO one clearly stands as the best-performing one. Incorporating MO into the serine surfactant system affects the morphology and size distribution of the formed mixed aggregates. In the low concentration regime, gemini–MO systems aggregate in the form of vesicles, while at high concentrations the formation of a lamellar liquid crystalline phase is observed. This suggests that lipoplexes might share a similar bilayer-based structure. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Molecular structure of the serine-based bis-quat surfactants used in this work and monoolein (MO).</p>
Full article ">Figure 2
<p>Mean <span class="html-italic">ζ</span>-potential ± (<span class="html-italic">SD</span>) (<b>a</b>) and mean diameter ± (<span class="html-italic">SD</span>) (<b>b</b>) (<span class="html-italic">n</span> = 3) of lipoplexes of gemini–MO/DNA as a function of gemini/DNA charge ratio, CR (+/−). The total gemini+MO concentration is 1 mM and the gemini:MO molar ratio in the lipoplexes varies between (2:1) and (1:4).</p>
Full article ">Figure 3
<p>Percentage of complexed DNA for gemini:MO lipoplexes, at different gemini/DNA charge ratios, CR (+/−). (<b>a</b>) (12Ser)<sub>2</sub>N5:MO/DNA systems, (<b>b</b>) (14Ser)<sub>2</sub>N5:MO/DNA systems, (<b>c</b>) (16Ser)<sub>2</sub>N5:MO/DNA systems, and (<b>d</b>) (18Ser)<sub>2</sub>N5:MO/DNA systems.</p>
Full article ">Figure 4
<p>Representative phase penetration scans for (12Ser)<sub>2</sub>N5:MO (<b>A</b>,<b>B</b>) and (14Ser)<sub>2</sub>N5:MO (<b>C</b>,<b>D</b>) at molar ratios 2:1 and 1:4, respectively. Water is diffusing from right to left into the surfactant crystalline film. Legend: L<sub>1</sub>, isotropic solution phase; H<sub>1</sub>, hexagonal phase; L<sub>α</sub>, lamellar phase; cr, hydrated crystals. Scale bars: (<b>A</b>–<b>D</b>) 150 µm; (<b>C1</b>,<b>C2</b>) 80 µm; and (<b>D1</b>) 50 µm.</p>
Full article ">Figure 5
<p>Representative imaging of (12Ser)<sub>2</sub>N5:MO and (18Ser)<sub>2</sub>N5:MO vesicles by light microscopy, for different gemini:MO molar ratios, for 10 mM dispersions.</p>
Full article ">Figure 6
<p>Dependence of cell viability ± standard deviation (<span class="html-italic">SD</span>) (<span class="html-italic">n</span> = 3) with increasing gemini:MO concentration after 48 h of exposure, for different gemini:MO molar ratios, from (2:1), (1:1), (1:2) to (1:4). (<b>a</b>) (12Ser)<sub>2</sub>N5:MO systems, (<b>b</b>) (14Ser)<sub>2</sub>N5:MO systems, (<b>c</b>) (16Ser)<sub>2</sub>N5:MO systems, and (<b>d</b>) (18Ser)<sub>2</sub>N5:MO systems.</p>
Full article ">
20 pages, 2758 KiB  
Article
Development of a Ready-to-Use-Type RNA Vaccine Carrier Based on an Intracellular Environment-Responsive Lipid-like Material with Immune-Activating Vitamin E Scaffolds
by Jessica Anindita, Hiroki Tanaka, Ryotaro Oyama, Shinya Hagiwara, Daiki Shirane, Sakura Taneichi, Yuta Nakai, Kota Tange, Hiroto Hatakeyama, Yu Sakurai and Hidetaka Akita
Pharmaceutics 2023, 15(12), 2702; https://doi.org/10.3390/pharmaceutics15122702 - 29 Nov 2023
Cited by 1 | Viewed by 2418
Abstract
Because of its efficient and robust gene transfer capability, messenger RNA (mRNA) has become a promising tool in various research fields. The lipid nanoparticle (LNP) is considered to be a fundamental technology for an mRNA delivery system and has been used extensively for [...] Read more.
Because of its efficient and robust gene transfer capability, messenger RNA (mRNA) has become a promising tool in various research fields. The lipid nanoparticle (LNP) is considered to be a fundamental technology for an mRNA delivery system and has been used extensively for the development of RNA vaccines against SARS-CoV-2. We recently developed ssPalm, an environmentally responsive lipid-like material, as a component of LNP for mRNA delivery. In this study, a self-degradable unit (phenyl ester) that confers high transfection activity and an immune stimulating unit (vitamin E scaffold) for high immune activation were combined to design a material, namely, ssPalmE-Phe-P4C2, for vaccine use. To design a simple and user-friendly form of an RNA vaccine based on this material, a freeze-drying-based preparation method for producing a ready-to-use-type LNP (LNP(RtoU)) was used to prepare the LNPssPalmE-Phe. The optimization of the preparation method and the lipid composition of the LNPssPalmE-Phe(RtoU) revealed that dioleoyl-sn-glycero phosphatidylethanolamine (DOPE) was a suitable helper lipid for achieving a high vaccination activity of the LNPssPalmE-Phe(RtoU). Other findings indicated that to maintain particle properties and vaccination activity, a 40% cholesterol content was necessary. A single administration of the LNPssPalmE-Phe(RtoU) that contained mRNA-encoding Ovalbumin (mOVA-LNPssPalmE-Phe(RtoU)) demonstrated a significant suppression of tumor progression in a tumor-bearing mouse OVA-expressing cell line (E.G7-OVA). In summary, the LNPssPalmE-Phe(RtoU) is an easy-to-handle drug delivery system (DDS) for delivering mRNA antigens in immunotherapy. Full article
(This article belongs to the Special Issue Advances in the Development of mRNA Medicines and mRNA Vaccines)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of lipid components and schematic illustration of the experimental design. (<b>a</b>) The molecular structures of ssPalmE-Phe-P4C2, ssPalmE-P4C2, DOPE, DOPC, cholesterol, and DMG-PEG<sub>2000</sub> are shown. (<b>b</b>) Experimental design of LNP<sub>ssPalmE-Phe</sub>(RtoU) preparation; the empty LNPs were prepared through microfluidic mixing and then lyophilized to produce dry empty LNPs. The mRNA-encapsulated LNPs could be obtained via the reconstitution (rehydration) of the dry LNPs with the mRNA solution in water.</p>
Full article ">Figure 2
<p>In vitro and in vivo gene expressions of LNPs<sub>ssPalmE</sub> and LNPs<sub>ssPalmE-Phe</sub> prepared with a microfluidic device. (<b>a</b>) Physicochemical properties of LNPs<sub>ssPalmE</sub> and LNPs<sub>ssPalmE-Phe</sub>. In vitro gene expression after transfection of mLuc-LNPs at a dose of 0.8 µg of mRNA in (<b>b</b>) BMDCs (8.0 × 10<sup>5</sup> cells) and (<b>c</b>) RAW 264.7 cells (2.0 × 10<sup>5</sup> cells). Luciferase activity is represented as a relative light unit (RLU/mg protein), calculated from the luminescence intensities and protein content of the cell lysates. (<b>d</b>,<b>e</b>) Quantification and imaging results of the in vivo mLuc-LNP gene expression in C57BL/6J mice through immunization (s.c.) of 1.0 µg of mRNA. Six hours after administration, D-luciferin potassium (3 mg/200 µL/mouse) was administered (i.p.), and the luminescence intensities were measured using an IVIS device. The scatter graph represents the individual value; the bar graph represents the mean with SD (<span class="html-italic">n</span> = 3); * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 (two-tail unpaired <span class="html-italic">t</span>-test). The measurements in the in vitro experiments were conducted in triplicate.</p>
Full article ">Figure 3
<p>CTL activity of the LNPs<sub>ssPalmE</sub> and LNPs<sub>ssPalmE-Phe</sub> prepared using a microfluidic device. The mOVA-LNPs<sub>ssPalmE</sub> and mOVA-LNPs<sub>ssPalmE-Phe</sub>, with either DOPE or DOPC as helper lipids, were evaluated for their immune activation activity via a CTL assay. The CTL assays of the mOVA-LNPs were conducted in C57BL/6J mice via immunization (s.c.) of 0.05 µg of mRNA. On day 7 after immunization, the mice were injected (i.v.) with equal amounts of CFSE<sup>hi</sup>-labelled splenocytes (SIINFEKL OVA epitope) and CFSE<sup>low</sup>-labelled splenocytes (naïve). The spleen was collected on Day 8, and the % lysis of splenocytes was quantified via flow cytometry. The scatter graph represents individual values; the bar graph represents the mean with SD (<span class="html-italic">n</span> = 3). N.S.: not significant; * <span class="html-italic">p</span> &lt; 0.05 (one-way ANOVA followed by the Bonferroni test against the original LNP<sub>ssPalmE</sub>).</p>
Full article ">Figure 4
<p>CTL activity of LNPs<sub>ssPalmE-Phe</sub>(RtoU) with different lipid compositions. (<b>a</b>) The activation of the antigen-specific cellular immunity of the LNPs<sub>ssPalmE-Phe</sub>(RtoU) with the lipid composition of ssPalmE-Phe-P4C2/(DOPE or DOPC)/cholesterol = 60/30/10 and ssPalmE-Phe-P4C2/(DOPE or DOPC)/cholesterol = 52.5/7.5/40 was evaluated via CTL assays. All lipid compositions included an additional 3 mol% of DMG-PEG<sub>2000</sub>. (<b>b</b>) The overlaid (left) and offset (right) FACS histogram from the CTL assay. CTL assays of mOVA-LNPs(RtoU) were conducted in C57BL/6J mice via immunization (s.c.) with 0.1 µg of mRNA. The spleen was collected, and the % lysis of splenocytes was quantified via flow cytometry. The scatter graph represents the individual values; the bar graph represents the mean with SD (<span class="html-italic">n</span> = 3). N.S.: not significant; * <span class="html-italic">p</span> &lt; 0.05 (one-way ANOVA followed by SNK test).</p>
Full article ">Figure 5
<p>CTL activity of LNPs<sub>ssPalmE-Phe</sub>(RtoU) prepared using different buffer pHs and sucrose concentrations. The CTL activity for combined buffer pH(s) and sucrose concentrations was evaluated and exhibited acceptable appearances and physicochemical properties, as shown in <a href="#pharmaceutics-15-02702-t002" class="html-table">Table 2</a>. A CTL assay of mOVA-LNPs(RtoU) was conducted in C57BL/6J mice via immunization (s.c.) with 0.1 µg of mRNA. The spleen was collected, and the % lysis of splenocytes was quantified via flow cytometry. The scatter graph represents individual values; the bar graph represents the mean with SD (<span class="html-italic">n</span> = 3). N.S.: not significant; ** <span class="html-italic">p</span> &lt; 0.01 (one-way ANOVA followed by SNK test).</p>
Full article ">Figure 6
<p>CTL activity evaluation of LNPs<sub>ssPalmE-Phe</sub>(RtoU) prepared using various lipid compositions. The LNPs<sub>ssPalmE-Phe</sub>(RtoU) were prepared with a total of 24 lipid compositions (<a href="#app1-pharmaceutics-15-02702" class="html-app">Table S2</a>), and their immune activation activities were evaluated via CTL assays at a dose of 0.1 µg of mRNA. (<b>a</b>) CTL activity of LNPs<sub>ssPalmE-Phe</sub>(RtoU) with 40% Chol. (<b>b</b>) CTL activity of LNPs<sub>ssPalmE-Phe</sub>(RtoU) with 0–10% DOPE (with 40% Chol). (<b>c</b>) CTL activity of LNPs<sub>ssPalmE-Phe</sub>(RtoU) with 0–10% DOPE (with 50% Chol). (<b>d</b>) CTL activity of LNPs<sub>ssPalmE-Phe</sub>(RtoU) with a DOPE ratio of 5–10% and 40 or 50% Chol. The lipid composition of ssPalmE-Phe-P4C2/DOPE/Chol = 52.5/7.5/40 was still found to be the favorable formulation for LNPs<sub>ssPalmE-Phe</sub>(RtoU). The scatter graph represents individual values; the bar graph represents the mean with SD (<span class="html-italic">n</span> = 3); white bar: control group; N.S.: not significant; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 (one-way ANOVA followed by Bonferroni testing against the control group). There were no significant differences found in all groups (against control group) in <a href="#pharmaceutics-15-02702-f006" class="html-fig">Figure 6</a>a.</p>
Full article ">Figure 7
<p>Therapeutic anti-tumor response of LNPs<sub>ssPalmE-Phe</sub>(RtoU). The antitumor response of LNPs<sub>ssPalmE-Phe</sub>(RtoU) against E.G7-OVA tumor-bearing mice was evaluated. E.G7-OVA cells (8.0 × 10<sup>5</sup> cells/40 µL) were s.c. inoculated on the left flank of C57BL/6J mice (<span class="html-italic">n</span> = 5). After the tumor grew to ≥100 mm<sup>3</sup> in volume, mOVA-LNP<sub>ssPalmE-Phe</sub>(RtoU) was administered (s.c.) at a dose of 0.5 or 1.0 µg of mRNA. The tumor volumes were monitored in 3-day intervals from day 9 to day 25. The end point was set at a tumor volume of 1000 mm<sup>3</sup>. (<b>a</b>) Individual tumor growth in each treatment group. (<b>b</b>) The overall survival of tumor-bearing mice was determined through a Kaplan–Meier analysis. Significant differences were found between mOVA-LNPs(RtoU), 0.5 µg and 1.0 µg, with PBS (* <span class="html-italic">p</span> = 0.01) and mLuc-LNPs(RtoU) (* <span class="html-italic">p</span> = 0.03).</p>
Full article ">
17 pages, 886 KiB  
Review
T Cells in Atherosclerosis: Key Players in the Pathogenesis of Vascular Disease
by Hannah Hinkley, Daniel A. Counts, Elizabeth VonCanon and Michael Lacy
Cells 2023, 12(17), 2152; https://doi.org/10.3390/cells12172152 - 26 Aug 2023
Cited by 15 | Viewed by 3357
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipid-rich plaques within arterial walls. T cells play a pivotal role in the pathogenesis of atherosclerosis in which they help orchestrate immune responses and contribute to plaque development and instability. Here, we [...] Read more.
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipid-rich plaques within arterial walls. T cells play a pivotal role in the pathogenesis of atherosclerosis in which they help orchestrate immune responses and contribute to plaque development and instability. Here, we discuss the recognition of atherosclerosis-related antigens that may trigger T cell activation together with additional signaling from co-stimulatory molecules and lesional cytokines. Although few studies have indicated candidates for the antigen specificity of T cells in atherosclerosis, further research is needed. Furthermore, we describe the pro-atherogenic and atheroprotective roles of diverse subsets of T cells such as CD4+ helper, CD8+ cytotoxic, invariant natural killer, and γδ T cells. To classify and quantify T cell subsets in atherosclerosis, we summarize current methods to analyze cellular heterogeneity including single cell RNA sequencing and T cell receptor (TCR) sequencing. Further insights into T cell biology will help shed light on the immunopathology of atherosclerosis, inform potential therapeutic interventions, and pave the way for precision medicine approaches in combating cardiovascular disease. Full article
Show Figures

Figure 1

Figure 1
<p>Role of T cell subsets in atherosclerosis. Lesional T cells can be broken up into three groups including T cells with α and β chains within their T cell receptor (TCR) like CD4<sup>+</sup> T cells (middle) and CD8<sup>+</sup> T cells (right) as well as non-conventional T cells that follow different activation pathways (left). Non-conventional T cells include the pro-atherogenic invariant natural killer T (iNKT) cells that express various T helper (T<sub>h</sub>) cytokines as well as interleukin (IL)-17 expressing γδ T cells. CD4<sup>+</sup> T cells include T<sub>h</sub> subsets such as pro-atherogenic T<sub>h</sub>1 that express interferon-γ, controversial T<sub>h</sub>2 that expresses IL-4 and IL-13, and T<sub>h</sub>17 that expresses IL-17. Follicular T helper (T<sub>FH</sub>) cells as well as CD28<sup>null</sup> T cells are both CD4<sup>+</sup>, which are pro-atherogenic. Additionally, regulatory T (Treg) are atheroprotective CD4<sup>+</sup> T cells; however, in late stage atherosclerosis, they convert to pro-atherogenic ex-Tregs. Finally, CD8<sup>+</sup> T cells can infiltrate atherosclerotic plaques as well, but their role remains unclear. Created with biorender.com.</p>
Full article ">
26 pages, 3610 KiB  
Article
Influence of the Composition of Cationic Liposomes on the Performance of Cargo Immunostimulatory RNA
by Ali Bishani, Darya M. Makarova, Elena V. Shmendel, Mikhail A. Maslov, Aleksandra V. Sen‘kova, Innokenty A. Savin, Daniil V. Gladkikh, Marina A. Zenkova and Elena L. Chernolovskaya
Pharmaceutics 2023, 15(9), 2184; https://doi.org/10.3390/pharmaceutics15092184 - 23 Aug 2023
Cited by 4 | Viewed by 2020
Abstract
In this study, the impact of different delivery systems on the cytokine-inducing, antiproliferative, and antitumor activities of short immunostimulatory double-stranded RNA (isRNA) was investigated. The delivery systems, consisting of the polycationic amphiphile 1,26-bis(cholest-5-en-3-yloxycarbonylamino)-7,11,16,20 tetraazahexacosan tetrahydrochloride (2X3), and the lipid-helper dioleoylphosphatidylethanolamine (DOPE), were equipped [...] Read more.
In this study, the impact of different delivery systems on the cytokine-inducing, antiproliferative, and antitumor activities of short immunostimulatory double-stranded RNA (isRNA) was investigated. The delivery systems, consisting of the polycationic amphiphile 1,26-bis(cholest-5-en-3-yloxycarbonylamino)-7,11,16,20 tetraazahexacosan tetrahydrochloride (2X3), and the lipid-helper dioleoylphosphatidylethanolamine (DOPE), were equipped with polyethylene glycol lipoconjugates differing in molecular weight and structure. The main findings of this work are as follows: (i) significant activation of MCP-1 and INF-α, β, and γ production in CBA mice occurs under the action of isRNA complexes with liposomes containing lipoconjugates with long PEG chains, while activation of MCP-1 and INF-γ, but not INF-α or β, was observed under the action of isRNA lipoplexes containing lipoconjugates with short PEG chains; (ii) a pronounced antiproliferative effect on B16 melanoma cells in vitro, as well as an antitumor and hepatoprotective effect in vivo, was induced by isRNA pre-complexes with non-pegylated liposomes, while complexes containing lipoconjugates with long-chain liposomes were inactive; (iii) the antitumor activity of isRNA correlated with the efficiency of its accumulation in the cells and did not explicitly depend on the activation of cytokine and interferon production. Thus, the structure of the delivery system plays a vital role in determining the response to isRNA and allows for the choice of a delivery system depending on the desired effect. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structures of cholesterol-based polycationic amphiphile 1,26-bis(cholest-5-en-3_-yloxycarbonylamino)-7,11,16,20-tetraazahexacosan tetrahydrochloride (2X3), lipid helper dioleoylphosphatidylethanolamine (DOPE), lipoconjugates: <span class="html-italic">O</span>,<span class="html-italic">O</span>′[<span class="html-italic">rac</span>-2,3-di(tetradecyloxy) propyl-1-oxycarbonylamino] octadecaethylene glycol (P800), <span class="html-italic">O</span>,<span class="html-italic">O</span>′-bis[<span class="html-italic">rac</span>-2,3-di(tetradecyloxy) propyl-1-oxycarbonylamino] octadecaethylene glycol (diP800), <span class="html-italic">O</span>,<span class="html-italic">O</span>′[<span class="html-italic">rac</span>-2,3-di(tetradecyloxy) propyl-1-oxycarbonylamino] poly(ethylene glycol<sub>1500</sub>) (P1500), <span class="html-italic">O</span>,<span class="html-italic">O</span>′-bis[<span class="html-italic">rac</span>-2,3-di(tetradecyloxy) propyl-1-oxycarbonylamino] poly(ethylene glycol<sub>1500</sub>) (diP1500), <span class="html-italic">O</span>,<span class="html-italic">O</span>′[<span class="html-italic">rac</span>-2,3-di(tetradecyloxy) propyl-1-oxycarbonylamino] poly(ethylene glycol<sub>2000</sub>) (P2000), <span class="html-italic">O</span>,<span class="html-italic">O</span>′-bis[<span class="html-italic">rac</span>-2,3-di(tetradecyloxy) propyl-1-oxycarbonylamino] poly(ethylene glycol<sub>2000</sub>) (diP2000) and <span class="html-italic">O</span>-{2-[rac-2,3-di(tetradecyloxy) prop-1-yloxycarbonyl]aminoethyl}-<span class="html-italic">O</span>′-[2-(pteroyl-L-glutam-5yl)aminoethyl]octadecaethyleneglycol (F12) is used for liposome preparation.</p>
Full article ">Figure 2
<p>Cytokine levels in human PBMC primary cell culture after cell transfection with isRNA/liposomes complexes. The levels of IFN-α (<b>A</b>) and TNF-α (<b>B</b>) 16 h after transfection and the levels IL-6 (<b>C</b>) 6 h after transfection were measured via ELISA. The data represent mean ± standard deviation (SD). Statistically significant differences between samples treated with isRNA/liposomes and the samples treated with empty liposomes are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.005); ordinary two-way ANOVA, Dunnett’s multiple comparisons test. Data for other time points are presented in <a href="#app1-pharmaceutics-15-02184" class="html-app">Supplementary Data (Figure S1)</a>.</p>
Full article ">Figure 2 Cont.
<p>Cytokine levels in human PBMC primary cell culture after cell transfection with isRNA/liposomes complexes. The levels of IFN-α (<b>A</b>) and TNF-α (<b>B</b>) 16 h after transfection and the levels IL-6 (<b>C</b>) 6 h after transfection were measured via ELISA. The data represent mean ± standard deviation (SD). Statistically significant differences between samples treated with isRNA/liposomes and the samples treated with empty liposomes are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.005); ordinary two-way ANOVA, Dunnett’s multiple comparisons test. Data for other time points are presented in <a href="#app1-pharmaceutics-15-02184" class="html-app">Supplementary Data (Figure S1)</a>.</p>
Full article ">Figure 3
<p>Cytokine production in CBA mice (<span class="html-italic">n</span> = 3) 6 h after i.v. administration of isRNA/liposomes complexes. (<b>A</b>) Serum IFN-α levels measured via ELISA. The data represent mean ± standard deviation (SD) calculated from samples from three mice measured in duplicates. Statistically significant differences between experimental groups and the control group are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.001); ordinary two-way ANOVA, Dunnett’s multiple comparisons test. (<b>B</b>) Cytokine profiling measured using LEGEND plex™ Mouse Inflammation Panel (13-plex) kit. Numerical data are presented in <a href="#app1-pharmaceutics-15-02184" class="html-app">Supplementary Data Table S1</a>.</p>
Full article ">Figure 4
<p>Antiproliferative activity of isRNA/liposomes on melanoma B16 and lymphosarcoma RLS40 tumor cells. (<b>A</b>) The experimental scheme. (<b>B</b>) Dynamics of lymphosarcoma RLS40 cell growth. (<b>C</b>) Dynamics of melanoma B16 cell growth. The relative number of living cells was assayed via WST-1; the number of cells at day 2 in control was set as 1. Statistically significant differences between experimental groups (isRNA/P2000 and P2000 in RLS40, isRNA/2X3-DOPE in B16) and control group are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.05 for P2000 and isRNA/P2000, ** <span class="html-italic">p</span> &lt; 0.005 for isRNA/2X3-DOPE); ordinary two-way ANOVA, Dunnett’s multiple comparisons test.</p>
Full article ">Figure 4 Cont.
<p>Antiproliferative activity of isRNA/liposomes on melanoma B16 and lymphosarcoma RLS40 tumor cells. (<b>A</b>) The experimental scheme. (<b>B</b>) Dynamics of lymphosarcoma RLS40 cell growth. (<b>C</b>) Dynamics of melanoma B16 cell growth. The relative number of living cells was assayed via WST-1; the number of cells at day 2 in control was set as 1. Statistically significant differences between experimental groups (isRNA/P2000 and P2000 in RLS40, isRNA/2X3-DOPE in B16) and control group are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.05 for P2000 and isRNA/P2000, ** <span class="html-italic">p</span> &lt; 0.005 for isRNA/2X3-DOPE); ordinary two-way ANOVA, Dunnett’s multiple comparisons test.</p>
Full article ">Figure 5
<p>Accumulation of Cy5.5-isRNA and its complexes with liposomes in B16 and RLS40 cells. The accumulation was measured 4 h after transfection of Cy5.5-isRNA mediated by liposomes using flow cytometry. Statistically significant differences between experimental groups and control group are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.005, ** <span class="html-italic">p</span> &lt; 0.0001); ordinary two-way ANOVA, Dunnett’s multiple comparisons test. RFU—Relative Fluorescence Unite.</p>
Full article ">Figure 6
<p>The levels of <span class="html-italic">Pkr</span> mRNA in RLS40 and B16 cells after treatment with isRNA/liposome or liposomes only. Relative expression of <span class="html-italic">Pkr</span> mRNA, normalized to the reference <span class="html-italic">Tbp</span> mRNA expression, was measured via RT-qPCR 24 h after transfection. <span class="html-italic">Pkr</span>/<span class="html-italic">Tbp</span> ratio in control was set as 1. Statistically significant differences between experimental groups and control group are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.005, ** <span class="html-italic">p</span> &lt; 0.0001); ordinary two-way ANOVA, Dunnett’s multiple comparisons test.</p>
Full article ">Figure 7
<p>Antitumor activity of isRNA/liposomes in the lymphosarcoma RLS40 and melanoma B16 mouse models. (<b>A</b>) Experimental setup. (<b>B</b>) Dynamics of tumor growth (<span class="html-italic">n</span> = 5 mice per group). (<b>C</b>) Average tumor weight at the end point of the experiment. The arrows indicate the day when mice received injections. Statistically significant differences between experimental groups and the control group are indicated by asterisks (* <span class="html-italic">p</span> ˂ 0.05 compared to control group, ** <span class="html-italic">p</span> &lt; 0.0005 for P2000 and isRNA/P2000 treated mice compared to the control group, *** <span class="html-italic">p</span> &lt; 0.0001 for 2X3-DOPE and isRNA/2X3-DOPE compared to the control group); ordinary two-way ANOVA, Dunnett’s multiple comparison test.</p>
Full article ">Figure 8
<p>The effect of isRNA/liposome on the liver tissue of B16 melanoma-bearing mice. Representative histological images of liver tissues of healthy animals, control, non-treated mice, mice that received isRNA/2X3-DOPE or isRNA/P2000 complexes, and mice treated with 2X3-DOPE or P2000 in OptiMEM, respectively. Insets show binuclear hepatocytes (<b>left</b>), necrosis in the liver parenchyma (red arrows), and hepatocytes with dystrophy (black arrows). Hematoxylin and eosin staining, original magnification ×400. Scale bar corresponds to 50 µm.</p>
Full article ">Figure 9
<p>The effect of isRNA/liposome on spleen tissue from mice with melanoma B16. (<b>A</b>) Morphometry data of spleen from C57BL\6 mice bearing melanoma B16. Volume density of white and red pulp in spleen tissue after 3 injections of isRNA/liposomes or liposomes only. (<b>B</b>) Representative histological images of the spleen sections. Hematoxylin and eosin staining. Black arrows indicate lymphoid follicles in the spleen. Original magnification ×100. The data represent mean ± standard deviation (SD) calculated from measurements from five mice in each group. Statistically significant differences between experimental groups and the control and healthy groups are indicated by asterisks (* = <span class="html-italic">p</span> value &lt; 0.001); ordinary two-way ANOVA, Dunnett’s multiple comparisons test. Scale bar corresponds to 200 µm.</p>
Full article ">
17 pages, 2183 KiB  
Article
Polysarcosine-Functionalized mRNA Lipid Nanoparticles Tailored for Immunotherapy
by Christoph Wilhelmy, Isabell Sofia Keil, Lukas Uebbing, Martin A. Schroer, Daniel Franke, Thomas Nawroth, Matthias Barz, Ugur Sahin, Heinrich Haas, Mustafa Diken and Peter Langguth
Pharmaceutics 2023, 15(8), 2068; https://doi.org/10.3390/pharmaceutics15082068 - 1 Aug 2023
Cited by 14 | Viewed by 4617
Abstract
Lipid nanoparticles (LNPs) have gained great attention as carriers for mRNA-based therapeutics, finding applications in various indications, extending beyond their recent use in vaccines for infectious diseases. However, many aspects of LNP structure and their effects on efficacy are not well characterized. To [...] Read more.
Lipid nanoparticles (LNPs) have gained great attention as carriers for mRNA-based therapeutics, finding applications in various indications, extending beyond their recent use in vaccines for infectious diseases. However, many aspects of LNP structure and their effects on efficacy are not well characterized. To further exploit the potential of mRNA therapeutics, better control of the relationship between LNP formulation composition with internal structure and transfection efficiency in vitro is necessary. We compared two well-established ionizable lipids, namely DODMA and MC3, in combination with two helper lipids, DOPE and DOPC, and two polymer-grafted lipids, either with polysarcosine (pSar) or polyethylene glycol (PEG). In addition to standard physicochemical characterization (size, zeta potential, RNA accessibility), small-angle X-ray scattering (SAXS) was used to analyze the structure of the LNPs. To assess biological activity, we performed transfection and cell-binding assays in human peripheral blood mononuclear cells (hPBMCs) using Thy1.1 reporter mRNA and Cy5-labeled mRNA, respectively. With the SAXS measurements, we were able to clearly reveal the effects of substituting the ionizable and helper lipid on the internal structure of the LNPs. In contrast, pSar as stealth moieties affected the LNPs in a different manner, by changing the surface morphology towards higher roughness. pSar LNPs were generally more active, where the highest transfection efficiency was achieved with the LNP formulation composition of MC3/DOPE/pSar. Our study highlights the utility of pSar for improved mRNA LNP products and the importance of pSar as a novel stealth moiety enhancing efficiency in future LNP formulation development. SAXS can provide valuable information for the rational development of such novel formulations by elucidating structural features in different LNP compositions. Full article
(This article belongs to the Special Issue Nanoparticles and Microparticles in Drug Delivery)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Small-angle X-ray scattering (SAXS) investigation. (<b>A</b>) SAXS patterns of different LNP formulations in phosphate buffer (pH 4.5, light blue) and application buffer (pH 5.7, dark blue). Formulations are displayed according to their composition with PEG-grafted LNPs (left) and pSar-grafted LNPs (right) and their ionizable and helper lipid are displayed right to the scattering patterns. Scattering patterns are vertically shifted for better visualization. (<b>B</b>) Comparison between LNP formulations in d-spacing (top). Investigated pairs generated with formulations only differing in one lipid component (ionizable lipid, helper lipid, stealth lipid). Mean of the differences in the compared formulations is shown on the right and represents the mean factor in which the formulations differ in d-spacing when comparing the investigated pairs; the same procedure for correlation length is at the bottom. Data displayed as mean ± S.D.</p>
Full article ">Figure 2
<p>In vitro tolerability of LNP formulation 1–8 in human peripheral blood mononuclear cells (hPBMC). Dose ranged from 100 ng to 2000 ng. Viability of each LNP formulation is shown as %Viable hPBMC. Data are presented as mean ± S.D., <span class="html-italic">n</span> = 3 technical replicates per LNP formulation.</p>
Full article ">Figure 3
<p>In vitro dose-dependent transfection efficiency of Monocytes for LNP formulations 1–8. Dose ranged from 100 ng to 2000 ng. Transfection efficiency of each LNP formulation is shown as %Thy1.1+ Monocytes. Data are presented as mean ± S.D., <span class="html-italic">n</span> = 3 technical replicates per LNP formulation.</p>
Full article ">Figure 4
<p>In vitro transfection efficiency of Thy1.1 RNA containing LNPs at a dose of 1000 ng in hPBMC, Monocytes as representative cell group. (<b>A</b>) Thy1.1-expressing Monocytes analyzed by flow cytometry. Numbers indicate the percentage of Thy1.1+ Monocytes. (<b>B</b>) Transfection efficiency of all PEG-lipid versus pSar-lipid LNPs shown as %Thy1.1+ Monocytes. Data are presented as mean ± S.D., analyzed by a two-way ANOVA with Šidák’s multiple comparison test, **** <span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">n</span> = 3 technical replicates per LNP formulation.</p>
Full article ">Figure 5
<p>In vitro cell-binding studies of Cy5-labeled RNA containing DODMA-LNP at a dose of 1000 ng in hPBMCs. (<b>A</b>) Cy5-labeled RNA-positive Monocytes analyzed by flow cytometry. Numbers indicate the percentage of Cy5+ Monocytes. (<b>B</b>) Cell-binding efficiency of each DODMA-LNP formulation is shown as %Cy5+ Monocytes. Data are presented as mean ± S.D., analyzed by a two-way ANOVA with Šidák’s multiple comparison test, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, <span class="html-italic">n</span> = 3 technical replicates per LNP formulation.</p>
Full article ">
14 pages, 850 KiB  
Review
Overview of Solid Lipid Nanoparticles in Breast Cancer Therapy
by Kyumin Mo, Ayoung Kim, Soohyun Choe, Miyoung Shin and Hyunho Yoon
Pharmaceutics 2023, 15(8), 2065; https://doi.org/10.3390/pharmaceutics15082065 - 31 Jul 2023
Cited by 9 | Viewed by 2592
Abstract
Lipid nanoparticles (LNPs), composed of ionized lipids, helper lipids, and cholesterol, provide general therapeutic effects by facilitating intracellular transport and avoiding endosomal compartments. LNP-based drug delivery has great potential for the development of novel gene therapies and effective vaccines. Solid lipid nanoparticles (SLNs) [...] Read more.
Lipid nanoparticles (LNPs), composed of ionized lipids, helper lipids, and cholesterol, provide general therapeutic effects by facilitating intracellular transport and avoiding endosomal compartments. LNP-based drug delivery has great potential for the development of novel gene therapies and effective vaccines. Solid lipid nanoparticles (SLNs) are derived from physiologically acceptable lipid components and remain robust at body temperature, thereby providing high structural stability and biocompatibility. By enhancing drug delivery through blood vessels, SLNs have been used to improve the efficacy of cancer treatments. Breast cancer, the most common malignancy in women, has a declining mortality rate but remains incurable. Recently, as an anticancer drug delivery system, SLNs have been widely used in breast cancer, improving the therapeutic efficacy of drugs. In this review, we discuss the latest advances of SLNs for breast cancer treatment and their potential in clinical use. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic drug loading models of SLNs; solid solution model, drug-enriched shell model, and drug-enriched core model. The solid solution model mainly consists of a dispersed drug in the solid lipid matrix and also has a high lipophilic interaction. In a drug-enriched shell model, the encapsulated drug is located in the outer shell and conversely, the drug-enriched core model shows the concentrated drug in the core. These drug distribution models are distinguished by their preparation methods and can be chosen for specific purposes in terms of size, characteristics, and release of the drug.</p>
Full article ">Figure 2
<p>Role of SLN-tamoxifen in tamoxifen-resistant cancer. Tamoxifen induces apoptosis of cancer cells via activating P-glycoprotein (P-gp) and inhibiting complex I-III in mitochondria. Complex I–III are mitochondria electron transport complex mediating ATP synthesis. However, P-gp is also known as a representative drug efflux transporter. In tamoxifen-resistant cancer cells, P-gp overexpression enhances drug efflux activity rather than the antitumor effect. SLN-tamoxifen overcomes resistance by delivering drugs directly to cytosol and inhibiting expression of anti-apoptotic genes.</p>
Full article ">
22 pages, 1427 KiB  
Review
A Review on the Involvement of Heat Shock Proteins (Extrinsic Chaperones) in Response to Stress Conditions in Aquatic Organisms
by Sivakamavalli Jeyachandran, Hethesh Chellapandian, Kiyun Park and Ihn-Sil Kwak
Antioxidants 2023, 12(7), 1444; https://doi.org/10.3390/antiox12071444 - 18 Jul 2023
Cited by 27 | Viewed by 5037
Abstract
Heat shock proteins (HSPs) encompass both extrinsic chaperones and stress proteins. These proteins, with molecular weights ranging from 14 to 120 kDa, are conserved across all living organisms and are expressed in response to stress. The upregulation of specific genes triggers the synthesis [...] Read more.
Heat shock proteins (HSPs) encompass both extrinsic chaperones and stress proteins. These proteins, with molecular weights ranging from 14 to 120 kDa, are conserved across all living organisms and are expressed in response to stress. The upregulation of specific genes triggers the synthesis of HSPs, facilitated by the interaction between heat shock factors and gene promoter regions. Notably, HSPs function as chaperones or helper molecules in various cellular processes involving lipids and proteins, and their upregulation is not limited to heat-induced stress but also occurs in response to anoxia, acidosis, hypoxia, toxins, ischemia, protein breakdown, and microbial infection. HSPs play a vital role in regulating protein synthesis in cells. They assist in the folding and assembly of other cellular proteins, primarily through HSP families such as HSP70 and HSP90. Additionally, the process of the folding, translocation, and aggregation of proteins is governed by the dynamic partitioning facilitated by HSPs throughout the cell. Beyond their involvement in protein metabolism, HSPs also exert a significant influence on apoptosis, the immune system, and various characteristics of inflammation. The immunity of aquatic organisms, including shrimp, fish, and shellfish, relies heavily on the development of inflammation, as well as non-specific and specific immune responses to viral and bacterial infections. Recent advancements in aquatic research have demonstrated that the HSP levels in populations of fish, shrimp, and shellfish can be increased through non-traumatic means such as water or oral administration of HSP stimulants, exogenous HSPs, and heat induction. These methods have proven useful in reducing physical stress and trauma, while also facilitating sustainable husbandry practices such as vaccination and transportation, thereby offering health benefits. Hence, the present review discusses the importance of HSPs in different tissues in aquatic organisms (fish, shrimp), and their expression levels during pathogen invasion; this gives new insights into the significance of HSPs in invertebrates. Full article
(This article belongs to the Special Issue Oxidative Stress and Antioxidant Response in Marine Organisms)
Show Figures

Figure 1

Figure 1
<p>Redox signaling mechanisms and inhibitory effects of HSPs in fish.</p>
Full article ">Figure 2
<p>Expression and molecular mechanisms of HSP and HSF in Mollusca.</p>
Full article ">Figure 3
<p>Types of heat shock proteins involved in folding and misfolding mechanisms.</p>
Full article ">
18 pages, 3398 KiB  
Article
HER2/neu Oncogene Silencing in a Breast Cancer Cell Model Using Cationic Lipid-Based Delivery Systems
by Adhika Balgobind, Aliscia Daniels, Mario Ariatti and Moganavelli Singh
Pharmaceutics 2023, 15(4), 1190; https://doi.org/10.3390/pharmaceutics15041190 - 8 Apr 2023
Cited by 9 | Viewed by 2063
Abstract
The overexpression of the human epidermal growth factor 2 (HER2/neu) oncogene is predictive of adverse breast cancer prognosis. Silencing the HER2/neu overexpression using siRNA may be an effective treatment strategy. Major requirements for siRNA-based therapy are safe, stable, and efficient delivery [...] Read more.
The overexpression of the human epidermal growth factor 2 (HER2/neu) oncogene is predictive of adverse breast cancer prognosis. Silencing the HER2/neu overexpression using siRNA may be an effective treatment strategy. Major requirements for siRNA-based therapy are safe, stable, and efficient delivery systems to channel siRNA into target cells. This study assessed the efficacy of cationic lipid-based systems for the delivery of siRNA. Cationic liposomes were formulated with equimolar ratios of the respective cholesteryl cytofectins, 3β-N-(N′, N′-dimethylaminopropyl)-carbamoyl cholesterol (Chol-T) or N, N-dimethylaminopropylaminylsuccinylcholesterylformylhydrazide (MS09), with the neutral helper lipid, dioleoylphosphatidylethanolamine (DOPE), with and without a polyethylene glycol stabilizer. All cationic liposomes efficiently bound, compacted, and protected the therapeutic siRNA against nuclease degradation. Liposomes and siRNA lipoplexes were spherical, <200 nm in size, with moderate particle size distributions (PDI < 0.4). The siRNA lipoplexes exhibited minimal dose-dependent cytotoxicity and effective HER2/neu siRNA transfection in the HER2/neu overexpressing SKBR-3 cells. The non-PEGylated Chol-T-siRNA lipoplexes induced the highest HER2/neu silencing at the mRNA (10000-fold decrease) and protein levels (>111.6-fold decrease), surpassing that of commercially available Lipofectamine 3000 (4.1-fold reduction in mRNA expression). These cationic liposomes are suitable carriers of HER2/neu siRNA for gene silencing in breast cancer. Full article
(This article belongs to the Special Issue Liposomal and Lipid-Based Drug Delivery Systems and Vaccines)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Transmission electron micrographs of cationic liposome: siRNA lipoplexes (N/P (+/−) charge ratios): (<b>A</b>) Chol-T; (<b>B</b>) Chol-T 2% PEG2000; (<b>C</b>) Chol-T 5% PEG2000; (<b>D</b>) MS09; (<b>E</b>) MS09 2% PEG22000; (<b>F</b>) MS09 5% PEG2000. Scale Bar = 100 nm (<b>D</b>) or 200 nm (<b>A</b>–<b>C</b>,<b>E</b>,<b>F</b>).</p>
Full article ">Figure 2
<p>Band-shift assay showing the binding interaction between varying amounts of PEGylated and non-PEGylated cationic liposomes with siRNA (0.32 μg). (<b>A</b>) Chol-T: Lanes 1–8 (0, 3.20, 3.52, 3.84, 4.16, 4.48, 4.80, and 5.12 μg); (<b>B</b>) Chol-T 2% PEG2000: Lanes 1–8 (0, 6.08, 6.40, 6.72, 7.04, 7.36, 7.68, and 8.00 μg); (<b>C</b>) Chol-T 5% PEG2000: Lanes 1–8 (0, 10.56, 10.88, 11.20, 11.52, 11.84, 12.16, and 12.48 μg); (<b>D</b>) MS09: Lanes 1–8 (0, 5.44, 5.76, 6.08, 6.40, 6.72, 7.04, and 7.36 μg); (<b>E</b>) MS09 2% PEG2000: Lanes 1–8 (0, 6.08, 6.40, 6.72, 7.04, 7.36, 7.68, and 8.00 μg); (<b>F</b>) MS09 5% PEG2000: Lanes 1–8 (0, 9.60, 9.92, 10.24, 10.56, 10.88, 11.20, and 11.52 μg). White arrows indicate endpoint ratios, except in 2F, where it indicates no clear endpoint.</p>
Full article ">Figure 3
<p>Nuclease protection assay of cationic and PEGylated cationic liposome-siRNA complexes in the presence of 10% FBS. Reaction mixtures (10 μL) contained siRNA (0.2 μg) and varying amounts of liposome suspension. (<b>A</b>) Lanes 3–5: Chol-T (2.4, 2.8, 3.2 μg) Lanes 6–8: MS09 (3.8, 4.2, 4.6 μg); (<b>B</b>) Lanes 3–5: Chol-T 2% PEG<sub>2000</sub> (4.4, 4.8, 5.2 μg), Lanes 6–8: Chol-T 5% PEG<sub>2000</sub> (7.0, 7.4, 7.8 μg); (<b>C</b>) Lanes 3–5: MS09 2% PEG<sub>2000</sub> (4.6, 5.0, 5.4 μg), Lanes 6–8: MS09 5% PEG<sub>2000</sub> (6.8, 7.2, 7.6 μg). Lane 1: FBS-untreated naked siRNA (0.2 μg) and lane 2: FBS-treated siRNA (0.2 μg).</p>
Full article ">Figure 4
<p>Cell viability studies of siRNA lipoplexes in (<b>A</b>) HEK293, (<b>B</b>) MCF-7, and (<b>C</b>) SKBR-3 cells in vitro. Incubation mixtures contained 0.32 μg of siRNA with varying amounts of liposome from suboptimal to supraoptimal N/P (+:−) charge ratios: Chol-T (3.4, 3.9, 4.4); Chol-T 2% PEG (5.8, 6.3, 6.8); Chol-T 5% PEG (8.3, 8.8, 9.3); MS09 (4.9, 5.4, 5.9); MS09 2% PEG (5.5, 6.0, 6.5); MS09 5% PEG (7.4, 7.9, 8.4). C1 = cells only- untreated), and C2 = Lipofectamine3000 treated cells. Data are presented as means ± SD (n = 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 were considered statistically significant vs. the cell control.</p>
Full article ">Figure 5
<p>Analysis of <span class="html-italic">HER2/neu</span> gene expression in SKBR-3 cells by qRT-PCR. The vertical axis represents the relative quantification of <span class="html-italic">HER2/neu</span> normalized against <span class="html-italic">GAPDH</span> mRNA level using the comparative quantification algorithm 2-∆∆Ct [<a href="#B30-pharmaceutics-15-01190" class="html-bibr">30</a>]. The mean ± SD of independent experiments (n = 3). *** <span class="html-italic">p</span> &lt; 0.001 was considered statistically significant.</p>
Full article ">Figure 6
<p>Analysis of HER2/<span class="html-italic">neu</span> oncoprotein expression by western blotting. (<b>A</b>) Non-treated SKBR-3 cells, NT-siRNA (non-targeting siRNA), and siRNA (<span class="html-italic">HER2/neu</span> targeting siRNA alone) served as negative controls. Lipofectamine<sup>®</sup> 3000-siRNA was included as a positive control, (<b>B</b>) SKBR-3 cells were treated with <span class="html-italic">HER2/neu</span> target siRNA: PEGylated and non-PEGylated Chol-T lipoplexes and (<b>C</b>) SKBR-3 cells were treated with HER2/<span class="html-italic">neu</span> target siRNA: PEGylated and non-PEGylated MS09 lipoplexes. HER2/<span class="html-italic">neu</span> receptor expression was determined in cellular lysates by Western blotting analysis using the HER2/<span class="html-italic">neu</span> and β-actin antibodies. Graphs represent the HER2/<span class="html-italic">neu</span>/β-actin normalization ratios. Data are presented as means ± SD (n = 3). **** <span class="html-italic">p</span> &lt; 0.0001 is considered statistically significant vs. the untreated SKBR-3 cell control.</p>
Full article ">Figure 7
<p>Comparison of HER2/neu oncogene knockdown of PEGylated and non-PEGylated (<b>A</b>) Chol-T and (<b>B</b>) MS09 lipoplexes vs. Lipofectamine<sup>®</sup> 3000 following Western Blotting Analysis. Graphs represent the HER2/neu/β-actin normalization ratios. Data are presented as means ± SD (n = 3). * <span class="html-italic">p</span> &lt; 0.1, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001 are considered statistically significant vs. Lipofectamine<sup>®</sup> 3000.</p>
Full article ">
15 pages, 9386 KiB  
Article
Efficient mRNA Delivery with mRNA Lipoplexes Prepared Using a Modified Ethanol Injection Method
by Min Tang, Ayane Sagawa, Nodoka Inoue, Satomi Torii, Kana Tomita and Yoshiyuki Hattori
Pharmaceutics 2023, 15(4), 1141; https://doi.org/10.3390/pharmaceutics15041141 - 4 Apr 2023
Cited by 8 | Viewed by 2792
Abstract
Messenger RNA (mRNA)-based therapies are a novel class of therapeutics used in vaccination and protein replacement therapies for monogenic diseases. Previously, we developed a modified ethanol injection (MEI) method for small interfering RNA (siRNA) transfection, in which cationic liposome/siRNA complexes (siRNA lipoplexes) were [...] Read more.
Messenger RNA (mRNA)-based therapies are a novel class of therapeutics used in vaccination and protein replacement therapies for monogenic diseases. Previously, we developed a modified ethanol injection (MEI) method for small interfering RNA (siRNA) transfection, in which cationic liposome/siRNA complexes (siRNA lipoplexes) were prepared by mixing a lipid-ethanol solution with a siRNA solution. In this study, we applied the MEI method to prepare mRNA lipoplexes and evaluated the in vitro and in vivo protein expression efficiencies. We selected six cationic lipids and three neutral helper lipids to generate 18 mRNA lipoplexes. These were composed of cationic lipids, neutral helper lipids, and polyethylene glycol-cholesteryl ether (PEG-Chol). Among them, mRNA lipoplexes containing N-hexadecyl-N,N-dimethylhexadecan-1-aminium bromide (DC-1-16) or 11-((1,3-bis(dodecanoyloxy)-2-((dodecanoyloxy)methyl) propan-2-yl) amino)-N,N,N-trimethyl-11-oxoundecan-1-aminium bromide (TC-1-12) with 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and PEG-Chol exhibited high protein expression in cells. Furthermore, mRNA lipoplexes composed of DC-1-16, DOPE, and PEG-Chol exhibited high protein expression in the lungs and spleen of mice after systemic injection and induced high antigen-specific IgG1 levels upon immunization. These results suggest that the MEI method can potentially increase the efficiency of mRNA transfection, both in vitro and in vivo. Full article
(This article belongs to the Special Issue Liposomal and Lipid-Based Drug Delivery Systems and Vaccines)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structure of cationic lipids, neutral helper lipids, and the PEG-lipid used in this study. DOTAP: 1,2-dioleoyl-3-trimethylammonium-propane methyl sulfate salt; DC-1-14: <span class="html-italic">N</span>,<span class="html-italic">N</span>-dimethyl-<span class="html-italic">N</span>-tetradecyltetradecan-1-aminium bromide; DC-1-16: <span class="html-italic">N</span>-hexadecyl-<span class="html-italic">N</span>,<span class="html-italic">N</span>-dimethylhexadecan-1-aminium bromide; DDAB: <span class="html-italic">N</span>,<span class="html-italic">N</span>-dimethyl-<span class="html-italic">N</span>-octadecyloctadecan-1-aminium bromide; DC-6-14: 2-(bis(2-(tetradecanoyloxy)ethyl)amino)-<span class="html-italic">N</span>,<span class="html-italic">N</span>,<span class="html-italic">N</span>-trimethyl-2-oxoethan-1-aminium chloride; TC-1-12: 11-((1,3-bis(dodecanoyloxy)-2-((dodecanoyloxy)methyl)propan-2-yl)amino)-<span class="html-italic">N</span>,<span class="html-italic">N</span>,<span class="html-italic">N</span>-trimethyl-11-oxoundecan-1-aminium bromide; DOPE: 1,2-dioleoyl-<span class="html-italic">sn</span>-glycero-3-phosphoethanolamine; DOPC: 1,2-dioleoyl-<span class="html-italic">sn</span>-glycero-3-phosphocholine; Chol: cholesterol; PEG-Chol: polyethylene glycol-cholesteryl ether.</p>
Full article ">Figure 2
<p>Effect of cationic and neutral helper lipids in mRNA lipoplexes on luciferase expression in HeLa cells after transfection with FLuc mRNA lipoplexes. mRNA lipoplexes with FLuc mRNA were added to HeLa cells at 0.5 μg/mL mRNA, and luciferase assays were carried out 24 h after incubation. Lipofectamine<sup>®</sup> MessengerMAX<sup>TM</sup> was used as a control for mRNA transfection. Each column represents the mean + S.D. (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Effect of cationic lipids in mRNA lipoplexes on EGFP expression in HeLa cells after transfection with EGFP mRNA lipoplexes. mRNA lipoplexes with EGFP mRNA were added to HeLa cells at 0.5 μg/mL mRNA, and EGFP expression (green) was observed 24 h after incubation. Scale bar = 100 μm. EGFP: enhanced green fluorescent protein.</p>
Full article ">Figure 4
<p>Effect of cationic lipids in mRNA lipoplexes on cellular uptake in HeLa cells after transfection with Cy5-mRNA lipoplexes. mRNA lipoplexes with Cy5-mRNA were added to HeLa cells at 0.5 μg/mL mRNA, and localization of Cy5-mRNA (green) was observed 3 h after incubation. Scale bar = 100 μm.</p>
Full article ">Figure 5
<p>Effect of incubation time and mRNA concentration on luciferase expression in HeLa cells after transfection with FLuc mRNA lipoplexes. LP-DC-1-14/DOPE, LP-DC-1-16/DOPE, and LP-TC-1-12/DOPE lipoplexes with FLuc mRNA were added to HeLa cells. (<b>A</b>) Luciferase activities were measured 24 h after transfection of mRNA lipoplexes at 0.5, 1, or 2 μg/mL mRNA. (<b>B</b>) Luciferase activities were measured 4, 24, or 48 h after transfection of mRNA lipoplexes with FLuc mRNA at 0.5 μg/mL mRNA. Each column represents the mean + S.D. (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 6
<p>Effect of cationic lipids in mRNA lipoplexes on cell viability 24 h after transfection of FLuc mRNA lipoplexes into HeLa cells. FLuc mRNA lipoplexes were added to HeLa cells at 0.5 μg/mL mRNA. Each column represents the mean + S.D. (<span class="html-italic">n</span> = 6). *** <span class="html-italic">p</span> &lt; 0.001, compared to untreated cells.</p>
Full article ">Figure 7
<p>Effect of cell types on luciferase expression and viability in A549 and PC-3 cells after transfection with FLuc mRNA lipoplexes. LP-DC-1-14/DOPE, LP-DC-1-16/DOPE, and LP-TC-1-12/DOPE lipoplexes with FLuc mRNA were added to A549 (<b>A</b>,<b>C</b>) and PC-3 cells (<b>B</b>,<b>D</b>) at 0.5 μg/mL mRNA, and luciferase activity (<b>A</b>,<b>B</b>) and cell viability (<b>C</b>,<b>D</b>) were measured 24 h after incubation. Each column represents the mean + S.D. (<span class="html-italic">n</span> = 3 for A and B, <span class="html-italic">n</span> = 6 for C and D). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared to untreated cells.</p>
Full article ">Figure 8
<p>mRNA biodistribution and protein expression in mice after IM injection of mRNA lipoplexes. (<b>A</b>) mRNA lipoplexes with 5 μg of Cy5-mRNA were intramuscularly injected into mice. The mice were sacrificed 1 or 24 h after injection, and Cy5 fluorescence images of the tissues were acquired with an exposure time of 5 s. Scale bar = 1 cm (<b>B</b>) mRNA lipoplexes with 5 μg of FLuc mRNA (5moU) were administered intramuscularly into mice. The mice were sacrificed 4 or 24 h after injection, and the luciferase activity of the tissues was measured. Each column represents the mean + S.D. (<span class="html-italic">n</span> = 4–6).</p>
Full article ">Figure 9
<p>mRNA biodistribution and protein expression in mice after IV injection of mRNA lipoplexes. (<b>A</b>) mRNA lipoplexes with 10 μg of Cy5-mRNA were intravenously injected into mice. The mice were sacrificed 1 h after injection, and Cy5 fluorescence images of the tissues were acquired with an exposure time of 5 s. Scale bar = 1 cm. (<b>B</b>) mRNA lipoplexes with 20 μg of FLuc mRNA (5moU) were administered systemically into mice. The mice were sacrificed 4 or 24 h after injection, and the luciferase activity of the tissues was measured. Each column represents the mean + S.D. (<span class="html-italic">n</span> = 4–5).</p>
Full article ">Figure 10
<p>Induction of anti-OVA IgG1 after IV injection of OVA mRNA lipoplexes. LP-DC-1-16/DOPE lipoplexes with 20 μg FLuc mRNA (5moU) or OVA mRNA (5moU) were systemically injected into the mice on days 0 and 14. Blood was collected from immunized mice on day 28, and OVA-specific IgG1 in serum was quantified. Each column represents the mean + S.D. (<span class="html-italic">n</span> = 4). OVA: ovalbumin.</p>
Full article ">
18 pages, 28740 KiB  
Review
Recent Advances in the Lipid Nanoparticle-Mediated Delivery of mRNA Vaccines
by K. Swetha, Niranjan G. Kotla, Lakshmi Tunki, Arya Jayaraj, Suresh K. Bhargava, Haitao Hu, Srinivasa Reddy Bonam and Rajendra Kurapati
Vaccines 2023, 11(3), 658; https://doi.org/10.3390/vaccines11030658 - 14 Mar 2023
Cited by 32 | Viewed by 10471
Abstract
Lipid nanoparticles (LNPs) have recently emerged as one of the most advanced technologies for the highly efficient in vivo delivery of exogenous mRNA, particularly for COVID-19 vaccine delivery. LNPs comprise four different lipids: ionizable lipids, helper or neutral lipids, cholesterol, and lipids attached [...] Read more.
Lipid nanoparticles (LNPs) have recently emerged as one of the most advanced technologies for the highly efficient in vivo delivery of exogenous mRNA, particularly for COVID-19 vaccine delivery. LNPs comprise four different lipids: ionizable lipids, helper or neutral lipids, cholesterol, and lipids attached to polyethylene glycol (PEG). In this review, we present recent the advances and insights for the design of LNPs, as well as their composition and properties, with a subsequent discussion on the development of COVID-19 vaccines. In particular, as ionizable lipids are the most critical drivers for complexing the mRNA and in vivo delivery, the role of ionizable lipids in mRNA vaccines is discussed in detail. Furthermore, the use of LNPs as effective delivery vehicles for vaccination, genome editing, and protein replacement therapy is explained. Finally, expert opinion on LNPs for mRNA vaccines is discussed, which may address future challenges in developing mRNA vaccines using highly efficient LNPs based on a novel set of ionizable lipids. Developing highly efficient mRNA delivery systems for vaccines with improved safety against some severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants remains difficult. Full article
(This article belongs to the Section Vaccines against Tropical and other Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>Composition of LNPs and importance of each component.</p>
Full article ">Figure 2
<p>Shows the different types of ionizable lipids used in the composition of LNPs.</p>
Full article ">Figure 3
<p>Shows the addition of a SORT lipid molecule to typical four-component LNPs alters the in vivo delivery profile of the resultant five-component SORT LNPs, allowing for tissue-specific distribution of mRNA to the liver, lungs, and spleen of mice following IV injections.</p>
Full article ">Figure 4
<p>(<b>A</b>). Structure and in vitro synthesis of mRNA. The basic structure of mRNA for therapeutics development. Cap, 5′ UTR, ORF, 3′ UTR, and PolyA are the five structural components of mRNA. (<b>B</b>). In vitro Synthesis of Modified mRNA. Mostly, vector DNA is used to construct the mRNA sequence. RNA polymerase uses the linearized DNA template containing either T7 or T3 or another promoter sequence for in vitro transcription (IVT) of mRNA. In some instances, caping and poly A addition can be done after the IVT process. However, some advanced kits are engineered to do the capping and poly A during the IVT process as a single step. More details are reviewed elsewhere [<a href="#B1-vaccines-11-00658" class="html-bibr">1</a>].</p>
Full article ">Figure 5
<p>Immune response by mRNA vaccines. LNPs are prepared by encapsulating mRNA, which encodes the viral protein of interest. Upon injection of vaccines, muscular cells take up the LNPs following the release of mRNA into the cytosol and translation of target protein with the help of host machinery. In parallel, the danger associated signals produced by the LNPs recruit the innate immune cells, including neutrophils, monocytes, macrophages, dendritic cells, and others. The antigen-presenting cells (APC) process and present the antigen to the T cells, which further polarizes into effector T cells and helps in B cell-mediated responses. The cytotoxic T cells produced upon activation kill the infected cells, and antibodies (produced by B cells or plasma cells) neutralize the virus.</p>
Full article ">Figure 6
<p>mRNA vaccine development. The figure illustrates the sequential steps involved in the mRNA vaccine development, from design to preclinical studies. Prior to going on to the next step, quality evaluation is required at each step.</p>
Full article ">
13 pages, 1667 KiB  
Communication
Influence of Cucurbiturils on the Production of Reactive Oxygen Species by T- and B-Lymphocytes, Platelets and Red Blood Cells
by Alina A. Aktanova, Olga S. Boeva, Margarita Sh. Barkovskaya, Ekaterina A. Kovalenko and Ekaterina A. Pashkina
Int. J. Mol. Sci. 2023, 24(2), 1441; https://doi.org/10.3390/ijms24021441 - 11 Jan 2023
Cited by 3 | Viewed by 1678
Abstract
Reactive oxygen species (ROS) are highly reactive chemical molecules containing oxygen. ROS play an important role in signaling and cell homeostasis at low and moderate concentrations. ROS could be a cause of damage to proteins, nucleic acids, lipids, membranes and organelles at high [...] Read more.
Reactive oxygen species (ROS) are highly reactive chemical molecules containing oxygen. ROS play an important role in signaling and cell homeostasis at low and moderate concentrations. ROS could be a cause of damage to proteins, nucleic acids, lipids, membranes and organelles at high concentrations. There are a lot of cells that can produce ROS to maintain functional activity. It is known that metal nanoparticles can increase production of ROS in cells. However, the effect of cucurbiturils on ROS production is still unknown. In our study, we evaluated production of ROS by the immune (T-, B-lymphocytes, NK-cells) and non-immune cells (red blood cells, platelets), as well as tumor cells line (1301, K562) after treatment with cucurbiturils in vitro. Assessment of reactive oxide species (ROS) were provided by using dihydrorhodamine 123 (DHR 123). Fluorescence intensity and percentage DHR123 were measured by flow cytometry. Platelets, erythrocytes and activated T-helpers were changed the level of ROS production in response to stimulation with cucurbiturils. It was found that the percentage of these ROS-producing cells was reduced by cucurbiturils. Thus, cucurbiturils may affect the production of ROS by cells, but further research is needed in this area. Full article
Show Figures

Figure 1

Figure 1
<p>Graph of the fluorescence intensity of erythrocytes, producing reactive oxygen species in response to stimulation with cucurbiturils (CB[6,7,8]) during 1 h. Data are presented as median ± interquartile range with n = 10. * <span class="html-italic">p</span> &lt; 0.05 by employing one-way ANOVA, Friedman test.</p>
Full article ">Figure 2
<p>Graph of the fluorescence intensity of platelets, producing reactive oxygen species in response to stimulation with cucurbiturils (CB [6,7,8]) during 1 h. Data are presented as median ± interquartile range with n = 10. * <span class="html-italic">p</span> &lt; 0.05 by employing one-way ANOVA, Friedman test.</p>
Full article ">Figure 3
<p>Graph of the percentage of cells of various subpopulations of lymphocytes, producing reactive oxygen species (<b>A</b>–<b>H</b>) in response to 24-h stimulation with cucurbiturils (CB [6,7,8]). Data are presented as median ± interquartile range with n = 10. * <span class="html-italic">p</span> &lt; 0.05 for comparing CB[6] group with control and PMA by employing one-way ANOVA, Friedman test.</p>
Full article ">Figure 4
<p>Graph of the amount of 1301 tumor cells (<b>A</b>) and the percentage of cells producing reactive oxygen species (<b>B</b>), Graph of the amount K562 tumor cells (<b>C</b>) and the percentage of cells producing reactive oxygen species (<b>D</b>) in response to 24-h stimulation with cucurbiturils (CB[6,7,8]). Data are median ± interquartile range (n = 6), (Kruskal–Wallis one-way analysis of variance). Notes: (<b>A</b>,<b>C</b>)—fluorescence intensity; (<b>B</b>,<b>D</b>)—number of ROS-producing cells among this cell’s population.</p>
Full article ">
13 pages, 3155 KiB  
Article
Influence of Lipid Composition of Cationic Liposomes 2X3-DOPE on mRNA Delivery into Eukaryotic Cells
by Vera Vysochinskaya, Sergey Shishlyannikov, Yana Zabrodskaya, Elena Shmendel, Sergey Klotchenko, Olga Dobrovolskaya, Nina Gavrilova, Darya Makarova, Marina Plotnikova, Ekaterina Elpaeva, Andrey Gorshkov, Dmitry Moshkoff, Mikhail Maslov and Andrey Vasin
Pharmaceutics 2023, 15(1), 8; https://doi.org/10.3390/pharmaceutics15010008 - 20 Dec 2022
Cited by 8 | Viewed by 3135
Abstract
The design of cationic liposomes for efficient mRNA delivery can significantly improve mRNA-based therapies. Lipoplexes based on polycationic lipid 1,26-bis(cholest-5-en-3β-yloxycarbonylamino)-7,11,16,20-tetraazahexacosane tetrahydrochloride (2X3) and helper lipid 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) were formulated in different molar ratios (1:1, 1:2, 1:3) to efficiently deliver model mRNAs to BHK-21 [...] Read more.
The design of cationic liposomes for efficient mRNA delivery can significantly improve mRNA-based therapies. Lipoplexes based on polycationic lipid 1,26-bis(cholest-5-en-3β-yloxycarbonylamino)-7,11,16,20-tetraazahexacosane tetrahydrochloride (2X3) and helper lipid 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) were formulated in different molar ratios (1:1, 1:2, 1:3) to efficiently deliver model mRNAs to BHK-21 and A549. The objective of this study was to examine the effect of 2X3-DOPE composition as well as lipid-to-mRNA ratio (amino-to-phosphate group ratio, N/P) on mRNA transfection. We found that lipoplex-mediated transfection efficiency depends on both liposome composition and the N/P ratio. Lipoplexes with an N/P ratio of 10/1 showed nanometric hydrodynamic size, positive ζ potential, maximum loading, and transfection efficiency. Liposomes 2X3-DOPE (1:3) provided the superior delivery of both mRNA coding firefly luciferase and mRNA-eGFP into BHK-21 cells and A549 cells, compared with commercial Lipofectamine MessengerMax. Full article
(This article belongs to the Special Issue Liposomal and Lipid-Based Drug Delivery Systems and Vaccines)
Show Figures

Figure 1

Figure 1
<p>Structural formula of the cationic lipid 2X3.</p>
Full article ">Figure 2
<p>TEM images of 2X3-DOPE 1:1, 2X3-DOPE 1:2, and 2X3-DOPE 1:3 liposomes in water. Scale bar is 500 nm (upper panel) or 100 nm (lower panel).</p>
Full article ">Figure 3
<p>Physicochemical characteristics of lipoplexes with mRNA-eGFP and mRNA-FLuc in different N/P ratios.</p>
Full article ">Figure 4
<p>Liposomes and mRNA complex formation. (<b>a</b>) Capillary electrophoresis method. Electropherograms of free mRNA (RNA) and lipoplexes at N/P ratios of 1/1, 2/1, and 4/1. (<b>b</b>) Ribogreen assay for quantification of encapsulated mRNA.</p>
Full article ">Figure 5
<p>(<b>a</b>) Surface topography of 2X3-DOPE 1:1, 2X3-DOPE 1:2, and 2X3-DOPE 1:3 liposomes without RNA, and the characteristic image of 2X3-DOPE complexes with mRNA (both eGFP and FLuc) at an N/P ratio of 10/1 (‘lipoplexes’). The scale bar is 1 µm. Pseudo-color rulers, reflecting the particle height distribution in each sample, are located on the right side of each image. The bottom row shows the characteristic particles of each sample in 3D view. (<b>b</b>) Diameter of the particles calculated based on AFM data. ‘ns’—not significant difference (<span class="html-italic">p</span>-value &gt; 0.05 based on one-way ANOVA statistics with correction for multiple comparisons using Tukey’s test); in all other cases, the samples significantly differ (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 6
<p>Transfection efficiency of mRNA using cationic liposomes 2X3-DOPE 1:1, 2X3-DOPE 1:2, and 2X3-DOPE 1:3 in BHK-21 cells 24 h after transfection. (<b>a</b>) Fluorescent microscopy results for mRNA-eGFP transfection at an N/P ratio of 10/1; green—eGFP protein; blue—cell nuclei. (<b>b</b>) Flow cytometry analysis of eGFP expression in transfected cells: the percentage of eGFP-positive cells (“transfected cells, %”, left) and the mean fluorescence intensity (MFI) of the cells (right) vs. the N/P ratio. (<b>c</b>) Transfection efficiency of FLuc-coding mRNA delivered by lipoplexes in different N/P ratios. Luciferase expression is expressed as relative light units (RLUs). MM is a positive control for mRNA transfection (Lipofectamine MessengerMAX reagent).</p>
Full article ">Figure 7
<p>Transfection efficiency of mRNA-eGFP using cationic liposomes 2X3-DOPE 1:1, 2X3-DOPE 1:2, and 2X3-DOPE 1:3 at an N/P ratio of 10/1 in A549 cells 24 h after transfection. (<b>a</b>) Fluorescent microscopy results; green—eGFP protein; blue—cell nuclei. (<b>b</b>) Flow cytometry analysis of eGFP expression in transfected cells: the percentage of eGFP-positive cells (“transfected cells, %”, left) and the mean fluorescence intensity (MFI) of the cells (right). (<b>c</b>) Transfection efficiency of FLuc-coding mRNA delivered by lipoplexes. Luciferase expression is expressed as relative light units (RLUs). Molar ratios of 1:1, 1:2, and 1:3 of 2X3 and DOPE, respectively. Lipofectamine MessengerMAX reagent (MM) is a positive control for mRNA transfection.</p>
Full article ">
17 pages, 3466 KiB  
Article
Effects of Different Routes and Forms of Vitamin D Administration on Mesenteric Lymph Node CD4+ T Cell Polarization and Intestinal Injury in Obese Mice Complicated with Polymicrobial Sepsis
by Chiu-Li Yeh, Jin-Ming Wu, Kuen-Yuan Chen, Ming-Hsun Wu, Po-Jen Yang, Po-Chu Lee, Po-Da Chen, Sung-Ling Yeh and Ming-Tsan Lin
Nutrients 2022, 14(17), 3557; https://doi.org/10.3390/nu14173557 - 29 Aug 2022
Cited by 6 | Viewed by 2554
Abstract
This study compared the efficacies of enteral cholecalciferol and/or intravenous (IV) calcitriol administration on mesenteric lymph node (MLN) cluster-of-differentiation-4-positive (CD4+) T cell distribution and intestinal barrier damage in obese mice complicated with sepsis. Mice were fed a high-fat diet for 16 weeks and [...] Read more.
This study compared the efficacies of enteral cholecalciferol and/or intravenous (IV) calcitriol administration on mesenteric lymph node (MLN) cluster-of-differentiation-4-positive (CD4+) T cell distribution and intestinal barrier damage in obese mice complicated with sepsis. Mice were fed a high-fat diet for 16 weeks and then sepsis was induced by cecal ligation and puncture (CLP). Mice were divided into the following sepsis groups: without vitamin D (VD) (S); with oral cholecalciferol 1 day before CLP (G); with IV calcitriol 1 h after CLP (V); and with both cholecalciferol before and IV calcitriol after CLP (GV). All mice were sacrificed at 12 or 24 h after CLP. The findings show that the S group had a higher T helper (Th)17 percentage than the VD-treated groups at 12 h after CLP. The V group exhibited a higher Th1 percentage and Th1/Th2 ratio than the other groups at 24 h, whereas the V and GV groups had a lower Th17/regulatory T (Treg) ratio 12 h post-CLP in MLNs. In ileum tissues, the VD-treated groups had higher tight junction protein and cathelicidin levels, and higher mucin gene expression than the S group at 24 h post-CLP. Also, aryl hydrocarbon receptor (AhR) and its associated cytochrome P450 1A1 and interleukin 22 gene expressions were upregulated. In contrast, levels of lipid peroxides and inflammatory mediators in ileum tissues were lower in the groups with VD treatment after CLP. These results suggest that IV calcitriol seemed to have a more-pronounced effect on modulating the homeostasis of Th/Treg subsets in MLNs. Both oral cholecalciferol before and IV calcitriol after CLP promoted cathelicidin secretion, alleviated intestinal inflammation, and ameliorated the epithelial integrity in obese mice complicated with sepsis possibly via VD receptor and AhR signaling pathways. Full article
(This article belongs to the Special Issue Regulatory Role of Vitamin D and Its Derivatives in the Immune System)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Concentrations of inflammatory mediators in peritoneal lavage fluid after cecal ligation and puncture (CLP) at two time points. IL-1β, interleukin-1β; IL-6, interleukin-6; TNF, tumor necrosis factor; MCP, macrophage chemoattractant protein; S, sepsis group without vitamin D (VD) before or after CLP; G, sepsis group with cholecalciferol gavage before CLP; V, sepsis group with an intravenous calcitriol injection after CLP; GV, sepsis group with both cholecalciferol before and intravenous calcitriol after CLP (<span class="html-italic">n</span> = 8 in each group). Data are presented as the mean ± standard error of the mean (SEM). Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Representative flow cytometry plots and percentages of T helper (Th) and regulatory T (Treg) cell subpopulations in mesenteric lymph nodes (MLNs) after cecal ligation and puncture at two time points. (<b>a</b>) Gating strategy for Th type 1 (Th1), Th2, and Th17. Lymphocytes were first identified based on Side Scatter (SSC) and Forward Scatter (FSC) characteristics. Cluster-of-differentiation-4-positive (CD4+) lymphocytes were gated to analyze the percentages of interferon (IFN)-γ-expressing, interleukin (IL)-4-expressing, and IL-17-expressing CD4 T cells. (<b>b</b>) Gating strategy for Treg cells. Forkhead box p3 (Foxp3)-expressing CD4+ lymphocytes were gated to identify the percentages of Treg cells. (<b>c</b>) Percentages of Th, Th2, Th17, and Treg cell subpopulations and the ratios of Th1/Th2 and Th17/Treg. The groups are described in the legend of <a href="#nutrients-14-03557-f001" class="html-fig">Figure 1</a> (<span class="html-italic">n</span> = 8 in each group). Data are presented as the mean ± SEM. Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point. <sup>+</sup> Significantly differs from the S and G groups at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2 Cont.
<p>Representative flow cytometry plots and percentages of T helper (Th) and regulatory T (Treg) cell subpopulations in mesenteric lymph nodes (MLNs) after cecal ligation and puncture at two time points. (<b>a</b>) Gating strategy for Th type 1 (Th1), Th2, and Th17. Lymphocytes were first identified based on Side Scatter (SSC) and Forward Scatter (FSC) characteristics. Cluster-of-differentiation-4-positive (CD4+) lymphocytes were gated to analyze the percentages of interferon (IFN)-γ-expressing, interleukin (IL)-4-expressing, and IL-17-expressing CD4 T cells. (<b>b</b>) Gating strategy for Treg cells. Forkhead box p3 (Foxp3)-expressing CD4+ lymphocytes were gated to identify the percentages of Treg cells. (<b>c</b>) Percentages of Th, Th2, Th17, and Treg cell subpopulations and the ratios of Th1/Th2 and Th17/Treg. The groups are described in the legend of <a href="#nutrients-14-03557-f001" class="html-fig">Figure 1</a> (<span class="html-italic">n</span> = 8 in each group). Data are presented as the mean ± SEM. Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point. <sup>+</sup> Significantly differs from the S and G groups at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Concentrations of inflammatory mediators in ileum tissues after cecal ligation and puncture at two time points. IL, interleukin; TNF, tumor necrosis factor; MCP, macrophage chemoattractant protein. The groups are described in the legend of <a href="#nutrients-14-03557-f001" class="html-fig">Figure 1</a> (<span class="html-italic">n</span> = 8 in each group). Data are presented as the mean ± SEM. Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Messenger (m)RNA expression levels of the vitamin D receptor (VDR) and cytochrome p450 27B1 (CYP27B1) genes in ileum tissues after cecal ligation and puncture at two time points. mRNA changes were quantitated and analyzed by real-time PCR and were calculated using the comparative CT (2<sup>−ΔΔCt</sup>) method. mRNA expression levels in the S group were used as a calibrator. Data are presented as the mean ± standard error of the mean (SEM). <span class="html-italic">n</span> = 8 for each group. The groups are described in the legend of <a href="#nutrients-14-03557-f001" class="html-fig">Figure 1</a>. Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Cathelicidin concentrations in ileum tissues after cecal ligation and puncture at two time points. Data are presented as the mean ± standard error of the mean (SEM). <span class="html-italic">n</span> = 8 for each group. The groups are described in the legend of <a href="#nutrients-14-03557-f001" class="html-fig">Figure 1</a>. Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Aryl hydrocarbon receptor (AhR), cytochrome P450 1A1 (CYP1A1), interleukin (IL)-22, and IL-22 receptor (IL-22R) mRNA levels in ileum tissues after cecal ligation and puncture at two time points. mRNA changes were quantitated and analyzed by real-time PCR and were calculated by the comparative CT (2<sup>−ΔΔCt</sup>) method. mRNA expression levels in the S group were used as a calibrator. The groups are described in the legend of <a href="#nutrients-14-03557-f001" class="html-fig">Figure 1</a>. Data are shown as the mean ± SEM (<span class="html-italic">n</span> = 8 for each group). Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point. <sup>+</sup> Significantly differs from the S and G groups at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 7
<p>(<b>a</b>) Tight junction protein levels and (<b>b</b>) mRNA levels of mucins in ileum tissues after cecal ligation and puncture at two time points. ZO-1, Zona occludens-1; Muc2, mucin 2; Tff3, trefoil factor 3. mRNA changes were quantitated and analyzed by real-time PCR and were calculated by the comparative CT (2<sup>−ΔΔCt</sup>) method. mRNA expression levels in the S group were used as a calibrator. The groups are described in the legend of <a href="#nutrients-14-03557-f001" class="html-fig">Figure 1</a>. Data are shown as the mean ± SEM (n = 8 for each group). Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point. <sup>+</sup> Significantly differs from the S and G groups at the same time point. <sup>‡</sup> Significantly differs from the S group at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 8
<p>Thiobarbituric-acid-reactive substance (TBARS) levels in ileum tissues after cecal ligation and puncture at two time points. The groups are described in the legend of <a href="#nutrients-14-03557-f001" class="html-fig">Figure 1</a> (n = 8 in each group). Values are expressed as the mean ± SEM. Differences among groups at the same time point were analyzed by a one-way analysis of variance using Tukey’s post hoc test. * Significantly differs from the other groups at the same time point (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
Back to TopTop