Nothing Special   »   [go: up one dir, main page]

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

Journals

Article Types

Countries / Regions

Search Results (22)

Search Parameters:
Keywords = hemoglobin vesicle

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 5382 KiB  
Article
Proteomic Analysis of Salivary Extracellular Vesicles from COVID-19 Patients Reveals a Specific Anti-COVID-19 Response Protein Signature
by Laura Weber, Alfredo Torres, Ornella Realini, María José Bendek, María Luisa Mizgier, Claudia Brizuela, David Herrera, Fermín E. González and Alejandra Chaparro
Int. J. Mol. Sci. 2024, 25(7), 3704; https://doi.org/10.3390/ijms25073704 - 26 Mar 2024
Cited by 1 | Viewed by 1708
Abstract
Despite the understanding of the coronavirus disease-19 (COVID-19), the role of salivary extracellular vesicles (sEVs) in COVID-19 remains unclear. Exploring the proteomic cargo of sEVs could prove valuable for diagnostic and prognostic purposes in assessing COVID-19. The proteomic cargo of sEVs from COVID-19(+) [...] Read more.
Despite the understanding of the coronavirus disease-19 (COVID-19), the role of salivary extracellular vesicles (sEVs) in COVID-19 remains unclear. Exploring the proteomic cargo of sEVs could prove valuable for diagnostic and prognostic purposes in assessing COVID-19. The proteomic cargo of sEVs from COVID-19(+) subjects and their healthy close contacts (HCC) was explored. sEVs were isolated by ultracentrifugation from unstimulated saliva samples, and subsequently characterized through nanoparticle tracking, transmission electron microscopy, and Western blot analyses. The proteomic cargo of sEVs was processed by LC-MS/MS. sEVs were morphologically compatible with EVs, with the presence of Syntenin-1 and CD81 EV markers. The sEV pellet showed 1417 proteins: 1288 in COVID-19(+) cases and 1382 in HCC. In total, 124 proteins were differentially expressed in sEVs from COVID-19(+) subjects. “Coronavirus-disease response”, “complement and coagulation cascades”, and “PMN extracellular trap formation” were the most enriched KEGG pathways in COVID-19(+) cases. The most represented biological processes were “Hemoglobin and haptoglobin binding” and “oxygen carrier activity”, and the best-denoted molecular functions were “regulated exocytosis and secretion” and “leucocyte and PMN mediated immunity”. sEV proteomic cargo in COVID-19(+) suggests activity related to immune response processes, oxygen transport, and antioxidant mechanisms. In contrast, in HCC, sEV signature profiles are mainly associated with epithelial homeostasis. Full article
(This article belongs to the Special Issue Exosomes and Extracellular Vesicles in Health and Diseases 2.0)
Show Figures

Figure 1

Figure 1
<p>Characterization of sEVs from COVID-19(+) and HCC subjects. (<b>A</b>) sEV distribution plot indicating size (nm) and concentration (particles/mL). (<b>B</b>) Bar plots for total, large, and small EVs in the HCC and COVID-19(+) groups. (<b>C</b>) Mode plot in the HCC and COVID-19(+) groups. (<b>D</b>) Size distribution and ratio of large/small plots for sEV subpopulations in the HCC and COVID-19(+) groups. (<b>E</b>) EV markers (Syntenin-1 and CD81) detected by WB in salivary EV and EV-depleted saliva. (<b>F</b>) Morphology of sEVs by TEM. Abbreviations: sEVs; salivary extracellular vesicles, HCC; healthy close contacts, WB; Western blot, TEM; transmission electron microscope.</p>
Full article ">Figure 2
<p>Overall protein profiles of the COVID-19(+) and HCC groups. (<b>A</b>) Venn diagram depicting overlapping proteins between the HCC and COVID-19(+) groups, including exclusive and more abundant (differentially expressed) proteins from each group. A total of 1417 proteins were identified, of which 124 were differentially expressed in the COVID-19(+) group. Among these, 35 were exclusive and 89 were more abundant. (<b>B</b>) Volcano plot: the log<sub>2</sub> (fold change) indicates the mean relative abundance for each protein. Each dot represents one single protein. The red and blue areas represent more abundant proteins with significant differences (<span class="html-italic">p</span> &lt; 0.05) in the COVID-19(+) and HCC groups, respectively. Dark colored dots represent hub proteins in the COVID-19(+) group. In contrast, the most abundant proteins in the HCC group are represented by dark colored dots, as their hub proteins were mainly exclusively found and were not represented in the plot. (<b>C</b>,<b>E</b>,<b>G</b>) Functional annotations and over-representation analysis of the top 10 most enriched KEGG pathways, GO molecular functions, and GO biological processes, respectively, of the exclusive and differentially expressed proteins (≥2-fold change, <span class="html-italic">p</span> &lt; 0.05) in the COVID-19(+) group. (<b>D</b>,<b>F</b>,<b>H</b>). Functional annotations and over-representation analysis of the top 10 most enriched KEGG pathways, GO molecular functions, and GO biological processes, respectively, of the exclusive and differentially expressed proteins (≥2-fold change, <span class="html-italic">p</span> &lt; 0.05) in the HCC group.</p>
Full article ">Figure 3
<p>Construction of PPI networks and identification of candidate hub proteins. (<b>A</b>,<b>C</b>). PPI network analysis in both the COVID-19(+) and HCC groups. In COVID-19(+), the PPI network consisted of 102 nodes and 281 edges, while in HCCs the PPI network consisted of 195 nodes and 610 edges, all of them were visualized in Cytoscape. A comprehensive view of the PPI network is illustrated in <a href="#app1-ijms-25-03704" class="html-app">Supplementary Figures S4 and S5</a> for both the COVID-19(+) and the HCC groups, respectively. (<b>B</b>,<b>D</b>) Top ten ranked proteins in network analysis with the MCC method in cytoHubba in COVID-19(+) and HCC, respectively. The top ten nodes are shown with a color scheme from red (highly important) to yellow (very important). (<b>E</b>) STRING PPI of hub proteins in the COVID-19(+) group: each color represents an enriched term. (<b>F</b>) KEGG pathway terms—the interactome network generated (<span class="html-italic">p</span> &lt; 0.005, FDR Q &lt; 0.05) of hub proteins from the COVID-19(+) group: each node represents an enriched pathway term. The nodes are connected by a line whose thickness reflects the percentage of overlapping proteins, and the size of the node corresponds to the number of proteins. Abbreviations: PPI, protein–protein interaction; MCC, maximal clique centrality.</p>
Full article ">Figure 4
<p>Functional interaction of the hub proteins. The chord diagram shows a detailed interaction of functional connections among the top 10 hub proteins (left side) from the HCC and COVID-19(+) groups and their assigned GO terms (right side).</p>
Full article ">Figure 5
<p>Schematic representation of differential ultracentrifugation-based sEV isolation and characterization methods in the context of symptomatic COVID-19 infection. Saliva samples were pooled based on clinical status (i.e., the COVID-19(+) and HCC groups). Differential ultracentrifugation-based extracellular vesicle isolation involved multiple cycles of centrifugation, ranging from 10,000× <span class="html-italic">g</span> up to 160,000× <span class="html-italic">g</span>. Following the final centrifugation, the supernatant was removed, and the enriched EV pellets were resuspended in 100 µL of DPBS. The concentration and distribution of sEVs were assessed using nano tracking particle analysis. Qualitative morphology assessment was conducted through transmission electron microscopy. EV markers (Syntenin-1, and CD81) and the contaminant marker (albumin) were evaluated by Western blot analysis. The proteomic cargo was determined by LC-MS/MS label-free analysis, and bioinformatics-assisted functional enrichment analyses were performed.</p>
Full article ">
9 pages, 1736 KiB  
Communication
Immunosuppressive Activity of Exosomes from Granulocytic Myeloid-Derived Suppressor Cells in a Murine Model of Immune Bone Marrow Failure
by Ash Lee Manley, Jichun Chen, Wendy Fitzgerald, Xingmin Feng and Neal S. Young
Int. J. Mol. Sci. 2023, 24(19), 14661; https://doi.org/10.3390/ijms241914661 - 28 Sep 2023
Cited by 1 | Viewed by 1437
Abstract
We previously reported that granulocytic myeloid-derived suppressor cells (G-MDSCs) suppressed T-cell activation and attenuated bone marrow failure (BMF) in a minor histocompatibility (minor-H) antigen mismatched murine aplastic anemia (AA) model. In the current study, we tested the hypothesis that exosomes, a subset of [...] Read more.
We previously reported that granulocytic myeloid-derived suppressor cells (G-MDSCs) suppressed T-cell activation and attenuated bone marrow failure (BMF) in a minor histocompatibility (minor-H) antigen mismatched murine aplastic anemia (AA) model. In the current study, we tested the hypothesis that exosomes, a subset of extracellular vesicles, are responsible at least partially for G-MDSCs’ therapeutic efficacy. Indeed, exosomes isolated from GMDSCs (G-MDSC-exos) suppressed CD4+ and CD8+ T-cell proliferation in vitro and mildly attenuated immune BMF in the minor-H mismatched AA model. G-MDSC-exos treatment significantly increased red blood cells, hemoglobin, and total bone marrow (BM) cells, and moderately reduced BM CD8+ T cells. G-MDSC-exos’ effects were associated with upregulations in an array of lymphocyte-suppression-related miRNAs such as hsa-miR-142-5p, miR-19a-3p, and miR-19b-3p in both BM CD4+ and CD8+ T cells. We concluded that G-MDSC-exos attenuate immune BMF via modulating the delivery of immunosuppressive miRNAs into activated T lymphocytes. Full article
(This article belongs to the Special Issue Molecular Immunology in Hematological Disorders 2.0)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Confirmation of exosomes using a nanoparticle tracking analyzer. Concentration and exosome size are depicted (<b>A</b>). The numbers in black represent the size of the exosome, corresponding to peaks in concentration. An image was taken of the exosomes during analysis using a nanoparticle tracking analyzer (<b>B</b>).</p>
Full article ">Figure 2
<p>Suppressive effects of G-MDSC exosomes on T-cell proliferation in vitro. (<b>A</b>) Preparation of GMDSC exosomes. G-MDSCs were isolated from the bone marrow of B6 mice and then cultured for 48 h at 37 °C with 5% CO<sub>2</sub> in complete RPMI1640 media for exosome release. The supernatant containing exosomes was cocultured with EXO-TC precipitation solution for 24 h at 4 °C to capture the exosomes. (<b>B</b>) Functional assay of exosomes on T-cell proliferation. Isolated exosomes from 20–30 × 10<sup>6</sup> BM G-MDSCs were cocultured with carboxyfluorescein succinimidyl ester (CFSE)-labeled B6 lymph node (LN) cells (1 × 10<sup>6</sup>/mL each reaction) stimulated with 50 ng/mL phorbol 12-myristate 13-acetate (PMA) and 500 µM ionomycin for 5 days in RPMI 1640 medium supplemented with 10% fetal calf serum and antibiotics. T-cell proliferation is indicated by the decrease in CFSE dye intensity as measured using median fluorescence intensity (MFI). The plots show one of two similar experiments. *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>G-MDSC exosomes attenuated immune-mediated bone marrow failure (BMF). (<b>A</b>) Male C.B10 mice were preirradiated with 5 Gys total body irradiation and infused with 5 × 10<sup>6</sup> lymph node (LN) cells/mouse from B6 male donors to induce BMF. Recipient mice were untreated (BMF, N = 20) or were treated with exosomes released from 12–18 × 10<sup>6</sup> B6 BM G-MDSCs following 48 h of culture in vitro (BMF + EXS, N = 14). (<b>B</b>) Mice were bled and euthanized on days 14–17 following LN-cell infusion, and peripheral blood was analyzed for neutrophils (NEUs), red blood cells (RBCs), hemoglobin (HGB), and platelets (PLTs), while the recovery of total BM cells was estimated based on marrow harvested from two tibiae and two femurs. (<b>C</b>) Proportions of BM CD4<sup>+</sup>, CD8<sup>+</sup> T cells, regulatory T cells (Treg) in CD4<sup>+</sup> T cells, and the ratio of Treg:CD8<sup>+</sup> T cells were measured via flow cytometry shown as representative dot plots and individual observation. (<b>D</b>) Proportions of apoptosis and viable residual bone marrow (RBM, excluding CD4<sup>+</sup> and CD8<sup>+</sup> T cells) were measured by flow cytometry shown as representative dot plots and individual observations. Data were combined from three separate experiments. (<b>E</b>) Cytokines in available plasma samples were measured by Luminex. *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Distinct miRNA profiles CD4<sup>+</sup> and CD8<sup>+</sup> T cells from exosome-treated bone marrow failure (BMF) mice. Scatter plots illustrating relative expression levels of 84 lymphocyte-activation-focused miRNAs in BM CD4<sup>+</sup> (<b>A</b>) and CD8<sup>+</sup> (<b>B</b>) T cells using pooled samples from BMF + EXS mice and BMF mice, respectively. The x-axis represents estimated expression differences measured in log10 for BMF control samples, and the y-axis represents estimated expression differences measured in log10 for BMF + EXS. Diagonal lines show a 2-fold expression difference between the two groups. Yellow dots indicate differentially expressed miRNAs in CD4<sup>+</sup> or CD8<sup>+</sup> T cells between two groups; red dots differentially expressed miRNAs overlapped between CD4<sup>+</sup> and CD8<sup>+</sup> T cells in two groups; black dots are miRNAs not differentially expressed between two groups. (<b>C</b>) Heat maps of differentially expressed miRNAs in CD4<sup>+</sup> (2 pools) and CD8<sup>+</sup> (3 pools) T cells between two groups. The red–blue color scale indicates normalized miRNA expression levels (red: high expression, blue: low expression). (<b>D</b>) The overlap of upregulated miRNAs between CD4<sup>+</sup> and CD8<sup>+</sup> T cells is represented using a Venn diagram.</p>
Full article ">
11 pages, 7270 KiB  
Article
Vascular Extracellular Vesicles Indicate Severe Hepatopulmonary Syndrome in Cirrhosis
by Sukriti Baweja, Anupama Kumari, Preeti Negi, Swati Thangariyal, P. Debishree Subudhi, Shivani Gautam, Ashmit Mittal and Chhagan Bihari
Diagnostics 2023, 13(7), 1272; https://doi.org/10.3390/diagnostics13071272 - 28 Mar 2023
Viewed by 1300
Abstract
Background: Hepatopulmonary syndrome (HPS) is a pulmonary vasculature complication in the setting of liver disease that is characterized by pathological vasodilation resulting in arterial oxygenation defects. We investigated the role of extracellular vesicles (EV) in cirrhosis patients with HPS, as well as the [...] Read more.
Background: Hepatopulmonary syndrome (HPS) is a pulmonary vasculature complication in the setting of liver disease that is characterized by pathological vasodilation resulting in arterial oxygenation defects. We investigated the role of extracellular vesicles (EV) in cirrhosis patients with HPS, as well as the functional effect of EV administration in a common bile duct ligation (CBDL) HPS mouse model. Methods: A total of 113 cirrhosis patients were studied: 42 (Gr. A) with HPS and 71 (Gr. B) without HPS, as well as 22 healthy controls. Plasma levels of EV associated with endothelial cells, epithelial cells, and hepatocytes were measured. The cytokine cargoes were estimated using ELISA. The effect of EV administered intranasally in the CBDL mouse model was investigated for its functional effect in vascular remodeling and inflammation. Results: We found endothelial cells (EC) associated EV (EC-EV) were elevated in cirrhosis patients with and without HPS (p < 0.001) than controls. EC-EV levels were higher in HPS patients (p = 0.004) than in those without HPS. The epithelial cell EVs were significantly high in cirrhosis patients than controls (p < 0.001) but no changes found in patients with HPS than without. There was a progressive increase in EC-EV levels from mild to severe intrapulmonary shunting in HPS patients (p = 0.02 mild vs. severe), and we were able to predict severe HPS with an AUROC of 0.85; p < 0.001. An inverse correlation of EC-EVs was found with hemoglobin (r = −0.24; p = 0.031) and PaO2 (r = 0.690; p = 0.01) and a direct correlation with MELD (r = 0.32; p = 0.014). Further, both TNF-α (p = 0.001) and IL-1β (p = 0.021) as cargo levels were significantly elevated inside the EVs of HPS patients than without HPS. Interestingly, upon administration of intranasal EVs, there was a significant decrease in Evans blue accumulation and lung wet–dry ratio (p = 0.042; 0.038). A significant reduction was also noticed in inflammation and cholestasis. Conclusion: High levels of plasma EC-EV levels were found in patients with HPS with elevated pro-inflammatory cytokine cargoes. EC-EVs were indicative of severe HPS condition. In the CBDL HPS model, we were able to prove the beneficial effects of improving vascular tone, inflammation, and liver pathogenesis. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Representative images from NanoSight NS300 show the quality, size, and concentration of total EVs isolated from plasma from healthy controls, patients with HPS, and patients without HPS. The Y-axis is concentration (particles/mL), and the X-axis is size in nm. (<b>B</b>–<b>D</b>) The dot plots show the absolute counts in EV/μL as measured using flow cytometry associated with endothelial cells (EC-EVs), epithelial cell EVs, and hepatocyte EVs. (<b>E</b>) The scatter plot shows the EC-EVs correlation with PaO2, MELD, and Hb.</p>
Full article ">Figure 2
<p>EC-EVs associated with severe intrapulmonary shunting. (<b>A</b>) The bar diagram depicts a significant increase in EC-EV levels in severe shunting versus in mild shunting (<span class="html-italic">p</span> = 0.00). (<b>B</b>) ROC analysis with estimation of the corresponding area under the curve (AUC) shows the predictive potential of EC-EVs for cirrhosis with HPS as mild vs. severe.</p>
Full article ">Figure 3
<p>Extracellular vesicle cargoes. (<b>A</b>) TNF-α levels inside plasma EVs of cirrhosis patients with and without HPS and healthy controls. (<b>B</b>) IL-1 β levels in EVs of cirrhosis patients. (<b>C</b>) IL-10 levels in EVs of cirrhosis patients.</p>
Full article ">Figure 4
<p>Extracellular vesicles (1 × 10<sup>8</sup> EV/μL) obtained from healthy WT mice were administered intranasally into CBDL mice (2 weeks post-surgery of CBDL) every day for 2 weeks. (<b>A</b>) The bar diagrams show the Evan blue accumulation in lungs and (<b>B</b>) liver/body weight in sham-operated and CBDL mice with and without EVs. (<b>C</b>) Bar graphs show the fold changes in mRNA candidate genes associated with vasodilators and inflammation.</p>
Full article ">Figure 5
<p>(<b>A</b>) Histology depicts ductular proliferation and inflammation, which were improved after intranasal administration of EVs. (<b>B</b>) Bar diagram shows the mRNA levels of candidate genes associated with hepatocytes, proliferation, inflammation, and apoptosis.</p>
Full article ">
13 pages, 1615 KiB  
Article
Methemoglobinemia, Increased Deformability and Reduced Membrane Stability of Red Blood Cells in a Cat with a CYB5R3 Splice Defect
by Sophia Jenni, Odette Ludwig-Peisker, Vidhya Jagannathan, Sandra Lapsina, Martina Stirn, Regina Hofmann-Lehmann, Nikolay Bogdanov, Nelli Schetle, Urs Giger, Tosso Leeb and Anna Bogdanova
Cells 2023, 12(7), 991; https://doi.org/10.3390/cells12070991 - 24 Mar 2023
Cited by 4 | Viewed by 2762
Abstract
Methemoglobinemia is an acquired or inherited condition resulting from oxidative stress or dysfunction of the NADH-cytochrome b5 reductase or associated pathways. This study describes the clinical, pathophysiological, and molecular genetic features of a cat with hereditary methemoglobinemia. Whole genome sequencing and mRNA transcript [...] Read more.
Methemoglobinemia is an acquired or inherited condition resulting from oxidative stress or dysfunction of the NADH-cytochrome b5 reductase or associated pathways. This study describes the clinical, pathophysiological, and molecular genetic features of a cat with hereditary methemoglobinemia. Whole genome sequencing and mRNA transcript analyses were performed in affected and control cats. Co-oximetry, ektacytometry, Ellman’s assay for reduced glutathione concentrations, and CYB5R activity were assessed. A young adult European domestic shorthair cat decompensated at induction of anesthesia and was found to have persistent methemoglobinemia of 39 ± 8% (reference range < 3%) of total hemoglobin which could be reversed upon intravenous methylene blue injection. The erythrocytic CYB5R activity was 20 ± 6% of normal. Genetic analyses revealed a single homozygous base exchange at the beginning of intron 3 of the CYB5R3 gene, c.226+5G>A. Subsequent mRNA studies confirmed a splice defect and demonstrated expression of two mutant CYB5R3 transcripts. Erythrocytic glutathione levels were twice that of controls. Mild microcytosis, echinocytes, and multiple Ca2+-filled vesicles were found in the affected cat. Erythrocytes were unstable at high osmolarities although highly deformable as follows from the changes in elongation index and maximal-tolerated osmolarity. Clinicopathological presentation of this cat was similar to other cats with CYB5R3 deficiency. We found that methemoglobinemia is associated with an increase in red blood cell fragility and deformability, glutathione overload, and morphological alterations typical for stress erythropoiesis. Full article
(This article belongs to the Collection Advances in Red Blood Cells Research)
Show Figures

Figure 1

Figure 1
<p>Microfluorescent live cell imaging of RBCs of the affected (metHb) and control cats. Bright field (BF) images of RBCs and Ca<sup>2+</sup> distribution (using fluorescent signal of Fluo-4 dye) from control and the affected cat. BF images of control (<b>A</b>,<b>B</b>) and the affected cat at first visit when the affected cat was anemic (<b>C</b>) and at second visit when in steady state (<b>D</b>). Panels E-H represent the Ca<sup>2+</sup>-dependent fluo-4 fluorescent signal for the RBCs of the control (<b>E</b>) and the affected (<b>F</b>–<b>H</b>) cats. Blue dotted frame indicates the BF and fluorescent images of the same area. Black arrow in panel C highlights an immature large RBC with distorted discoid shape. Platelets (<b>E</b>,<b>F</b>) are filled with Ca<sup>2+</sup> and intensely stained (green arrows). Some of the discocytes (blue arrows, (<b>F</b>,<b>H</b>)) and echinocytes (yellow arrows, (<b>H</b>)) of the affected cat were overloaded with Ca<sup>2+</sup>. A few Ca<sup>2+</sup>-overloaded cells of the affected cat contained multiple Ca<sup>2+</sup>-filled vesicles (<b>F</b>,<b>G</b>), which are characteristic for immature reticulocytes.</p>
Full article ">Figure 2
<p>Sanger sequencing electropherograms of the affected and a control cat illustrating the single nucleotide exchange at the beginning of intron 3. The variant position is indicated by an arrow. The altered base is indicated in red.</p>
Full article ">Figure 3
<p>Aberrant <span class="html-italic">CYB5R3</span> transcripts in the affected cat. (<b>A</b>) Fragment Analyzer bands of the RT–PCR products in the control animal show the expected 599 bp band, while in the affected animal, two different bands are visible (mut #1 and mut #2). Sanger sequencing yielded a length of 635 bp for mut #1 and 526 bp for mut #2. (<b>B</b>) Schematic representation of exons 2 to 4 of the <span class="html-italic">CYB5R3</span> gene. The c.226+5G&gt;A variant is indicated on the genomic DNA level with a blue arrow. The variant leads to aberrant splicing, adding 36 bp of intron 3 to the mut #1 mRNA and skipping the entire exon 3 in the mut #2 transcript.</p>
Full article ">Figure 4
<p>Protein modeling of CYB5R3. (<b>A</b>) Protein model of the wildtype feline CYB5R3 protein. (<b>B</b>) Protein model of the predicted translation product of transcript mut #1 (XP_044918404.1:p.G76_Q77insERSPDPARVEPG). The 12 inserted amino acids are marked in red. (<b>C</b>) Protein model of the predicted translation product of transcript mut #2 XP_044918404.1:p.(V52Afs*58). We did not experimentally verify whether the predicted proteins were expressed. However, the RT–PCR experiments confirmed that the mut #2 mRNA transcript was present in the affected cat, suggesting that at least a fraction of this aberrant transcript escaped nonsense-mediated mRNA decay.</p>
Full article ">
18 pages, 2845 KiB  
Article
Heterologous Display of Chlamydia trachomatis PmpD Passenger at the Surface of Salmonella OMVs
by Dung T. Huynh, Wouter S. P. Jong, Manon A. H. Oudejans, H. Bart van den Berg van Saparoea, Joen Luirink and Peter van Ulsen
Membranes 2023, 13(4), 366; https://doi.org/10.3390/membranes13040366 - 23 Mar 2023
Cited by 2 | Viewed by 2197
Abstract
Chlamydia trachomatis is the bacterial pathogen that causes most cases of sexually transmitted diseases annually. To combat the global spread of asymptomatic infection, development of effective (mucosal) vaccines that offer both systemic and local immune responses is considered a high priority. In this [...] Read more.
Chlamydia trachomatis is the bacterial pathogen that causes most cases of sexually transmitted diseases annually. To combat the global spread of asymptomatic infection, development of effective (mucosal) vaccines that offer both systemic and local immune responses is considered a high priority. In this study, we explored the expression of C. trachomatis full-length (FL) PmpD, as well as truncated PmpD passenger constructs fused to a “display” autotransporter (AT) hemoglobin protease (HbpD) and studied their inclusion into outer membrane vesicles (OMVs) of Escherichia coli and Salmonella Typhimurium. OMVs are considered safe vaccine vectors well-suited for mucosal delivery. By using E. coli AT HbpD-fusions of chimeric constructs we improved surface display and successfully generated Salmonella OMVs decorated with a secreted and immunogenic PmpD passenger fragment (aa68-629) to 13% of the total protein content. Next, we investigated whether a similar chimeric surface display strategy could be applied to other AT antigens, i.e., secreted fragments of Prn (aa35-350) of Bordetella pertussis and VacA (aa65-377) of Helicobacter pylori. The data provided information on the complexity of heterologous expression of AT antigens at the OMV surface and suggested that optimal expression strategies should be developed on an antigen-to-antigen basis. Full article
(This article belongs to the Special Issue Recent Advances in Outer Membrane Vesicles)
Show Figures

Figure 1

Figure 1
<p>Expression of <span class="html-italic">C. trachomatis</span> PmpD (FL) in different <span class="html-italic">E. coli</span> strains. (<b>A</b>) Schematic representation of the pLemo-PmpD (FL) construct. <span class="html-italic">E. coli</span> Hbp signal sequence (ssHbp) fused to <span class="html-italic">C. trachomatis</span> PmpD (FL) that consists of passenger and β-barrel domain under control of the <span class="html-italic">rhaBAD</span> promoter. (<b>B</b>,<b>C</b>) Whole-cell lysates of <span class="html-italic">E. coli</span> BL21(DE3) and BL21(DE3)omp8 cells induced with (+) or without (−) 8 mM L-rhamnose for 2 h for expression of PmpD (FL) were analyzed by SDS-PAGE, followed by Coomassie staining (<b>B</b>) or immunoblotting using PmpD antiserum (<b>C</b>). (<b>D</b>,<b>E</b>) A cellular subfractionation of BL21(DE3)omp8 cells expressing PmpD (FL) resulted in whole-cell lysate (WCL), cell envelope (CE) and outer membrane (OM) fractions. BL21(DE3)omp8 outer membrane vesicles (OMV) were collected from the filtered culture supernatant. Samples derived from 0.05 OD<sub>600</sub> units of WCL, CE, OM, as well as the equivalent 4 OD<sub>600</sub> units of <span class="html-italic">E. coli</span> OMVs were analyzed by SDS-PAGE followed by Coomassie staining (<b>D</b>) and immunoblotting against PmpD or LpoB antisera as indicated. In the panels (<b>B</b>–<b>E</b>), the proteolytic product of PmpD (FL) of ~120 kDa was annotated (*). MW markers (in kDa) are indicated at the left and identified protein bands at the right side of panels.</p>
Full article ">Figure 2
<p>Expression of HbpD(Δd1)-PmpD in different <span class="html-italic">E. coli</span> strains. (<b>A</b>) Schematic representation of the HbpD(Δd1)-PmpD (aa68-698) construct in which truncated PmpD (aa68-698) was fused to the HbpD(Δd1)-based platform and expressed under control of the <span class="html-italic">lacUV5</span> promoter. (<b>B</b>,<b>C</b>) BL21(DE3) and BL21(DE3)omp8 cells were induced for HbpD(Δd1)-PmpD expression with (+) or without (−) 0.1 mM IPTG for 2 h and whole cells were analyzed by SDS-PAGE/Coomassie staining (<b>B</b>) and immunoblotting using antiserum against PmpD (<b>C</b>). MW markers (in kDa) are indicated at the left and identified protein bands at the right side of panels.</p>
Full article ">Figure 3
<p>Display of HbpD(Δd1)-PmpD, HbpD(Δd1)-Prn and HbpD(Δd1)-VacA at the surface of <span class="html-italic">E. coli</span> OMVs. Equivalent amounts of BL21(DE3)omp8 OMVs expressing HbpD(Δd1)-PmpD; HbpD(Δd1)-Prn and HbpD(Δd1)-VacA were used for a proteinase K (ProK) accessibility assay. Samples of untreated intact OMVs, intact OMVs treated with ProK and lysed OMVs, treated with TritonX-100 (TX-100) and incubated with ProK were analyzed by SDS-PAGE followed by silver staining (<b>A</b>) or immunoblotting using Hbp antiserum (<b>B</b>) or antiserum recognizing the <span class="html-italic">E. coli</span> Bam complex (<b>C</b>). In the panel (<b>C</b>) bands representing truncated BamA were indicated (*). MW markers (in kDa) are indicated at the left and identified protein bands at the right side of panels.</p>
Full article ">Figure 4
<p>Expression of EV, HbpΔβcleavage, HbpD(Δd1)-PmpD, HbpD(Δd1)-Prn and HbpD(Δd1)-VacA in <span class="html-italic">Salmonella</span> and their OMVs. SL3261 Δ<span class="html-italic">tolRA</span> Δ<span class="html-italic">msbB</span> cells were induced for expression of empty vector pEH3 (EV), HbpΔβcleavage, HbpD(Δd1)-PmpD, HbpD(Δd1)-Prn and HbpD(Δd1)-VacA and whole cells were analyzed by SDS-PAGE followed by Coomassie staining (<b>A</b>) and immunoblotting using antisera against Hbp or DegP (<b>B</b>). Equivalent amounts of OMVs were analyzed by SDS-PAGE followed by Coomassie staining (<b>C</b>) or immunoblotting using Hbp antiserum (<b>D</b>). In the panel (<b>B</b>–<b>D</b>), bands representing proteolytically degraded HbpD(Δd1) were indicated (*). OMVs expressing HbpD(Δd1)-PmpD were used for a proteinase K (ProK) assay. Samples of untreated intact OMVs, intact OMVs treated with ProK, and lysed OMVs, treated with TritonX-100 (TX-100) and incubated with ProK were analyzed by SDS-PAGE followed by Coomassie staining (<b>E</b>) or immunoblotting using PmpD-directed antiserum (<b>F</b>). MW markers (in kDa) are indicated at the left and identified protein bands at the right side of panels.</p>
Full article ">Figure 5
<p>Expression of EV, HbpΔβcleavage, HbpD(840)-PmpD, HbpD(840)-Prn and HbpD(840)-VacA in <span class="html-italic">Salmonella</span> whole cell lysates and isolated <span class="html-italic">Salmonella</span> OMVs. SL3261 Δ<span class="html-italic">tolRA</span> Δ<span class="html-italic">msbB</span> cells were induced for expression in presence of the empty vector (EV) or the pEH3 plasmids encoding HbpΔβcleavage, HbpD(840)-PmpD, HbpD(840)-Prn and HbpD(840)-VacA. Whole cell lysates were analyzed by SDS-PAGE followed by Coomassie staining (<b>A</b>) or immunoblotting using antiserum against Hbp (<b>B</b>). Equivalent amounts of OMVs were analyzed by SDS-PAGE followed by Coomassie staining (<b>C</b>) or immunoblotting using antiserum against Hbp (<b>D</b>). OMVs expressing HbpD(840)-PmpD were used for a proteinase K (ProK) assay. Samples of untreated intact OMVs, intact OMVs treated with ProK and lysed OMVs, treated with TritonX-100 (TX-100) and incubated with ProK were analyzed by SDS-PAGE followed by Coomassie staining (<b>E</b>) or immunoblotting using Hbp antiserum (<b>F</b>). MW markers (in kDa) are indicated at the left and identified protein bands at the right side of panels.</p>
Full article ">
24 pages, 5297 KiB  
Review
Placenta-Derived Extracellular Vesicles in Pregnancy Complications and Prospects on a Liquid Biopsy for Hemoglobin Bart’s Disease
by Piya Chaemsaithong, Suchaya Luewan, Mana Taweevisit, Wararat Chiangjong, Pisut Pongchaikul, Paul Scott Thorner, Theera Tongsong and Somchai Chutipongtanate
Int. J. Mol. Sci. 2023, 24(6), 5658; https://doi.org/10.3390/ijms24065658 - 16 Mar 2023
Cited by 5 | Viewed by 3056
Abstract
Extracellular vesicles (EVs) are nano-scaled vesicles released from all cell types into extracellular fluids and specifically contain signature molecules of the original cells and tissues, including the placenta. Placenta-derived EVs can be detected in maternal circulation at as early as six weeks of [...] Read more.
Extracellular vesicles (EVs) are nano-scaled vesicles released from all cell types into extracellular fluids and specifically contain signature molecules of the original cells and tissues, including the placenta. Placenta-derived EVs can be detected in maternal circulation at as early as six weeks of gestation, and their release can be triggered by the oxygen level and glucose concentration. Placental-associated complications such as preeclampsia, fetal growth restriction, and gestational diabetes have alterations in placenta-derived EVs in maternal plasma, and this can be used as a liquid biopsy for the diagnosis, prediction, and monitoring of such pregnancy complications. Alpha-thalassemia major (“homozygous alpha-thalassemia-1”) or hemoglobin Bart’s disease is the most severe form of thalassemia disease, and this condition is lethal for the fetus. Women with Bart’s hydrops fetalis demonstrate signs of placental hypoxia and placentomegaly, thereby placenta-derived EVs provide an opportunity for a non-invasive liquid biopsy of this lethal condition. In this article, we introduced clinical features and current diagnostic markers of Bart’s hydrops fetalis, extensively summarize the characteristics and biology of placenta-derived EVs, and discuss the challenges and opportunities of placenta-derived EVs as part of diagnostic tests for placental complications focusing on Bart’s hydrop fetalis. Full article
(This article belongs to the Special Issue The Role of Exosomes in Health and Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Placental pathology in Hb Bart’s disease. (<b>a</b>) The placenta belonging to Hb Bart’s disease shows enlarged, pale, and edematous cut surfaces in comparison to the placenta of non-Hb Bart’s disease (<b>d</b>). (<b>b</b>) Immature intermediate villi with a bulbous contour possessing conspicuous myofibroblasts at the periphery beneath the trophoblastic layer referred to as peripheral villous stromal hypercellularity in comparison to the control (<b>e</b>) (hematoxylin and eosin, original magnification ×400). (<b>c</b>) A branching vascular pattern with increased vessel number and endothelial thickness in comparison to the control (<b>f</b>) (CD34 immunostaining, original magnification ×600).</p>
Full article ">Figure 2
<p>Extracellular vesicle biogenesis and compositions. (<b>a</b>) Exosome biogenesis and intracellular life are depicted on the left. Cell surface proteins are endocytosed and trafficked to early endosomes. Once sorted to late endosomes, the endosomal sorting complex required for transport of (ESCRT)-0 complex recruits ubiquitinated proteins, while ESCRT-I and -II mediate the budding of intraluminal vesicles (ILVs). The multivesicular body (MVB) can either follow a degradation pathway fusing with lysosomes or proceed to release the ILV contents (as exosomes) to the extracellular space by an exocytic step. Microvesicle biogenesis via plasma membrane budding is illustrated on the right. Transmembrane proteins are clustered in discrete membrane domains that promote outward membrane budding. Tetraspanins and other proteins abundant at the domain may have a role by promoting the sorting of other components. Lipid-anchored (myristoylation, palmitoylation) proteins accumulate proteins in the lumen as well as contribute to membrane curvature. Additional mechanisms of microvesicle formation include the calcium-activated scramblases, which randomize the distribution of lipids between the two faces of the plasma membrane. The cytoskeleton becomes looser, while cytosolic proteins and RNA molecules are sorted into microvesicles. The specific ATPase VPS4 mediates the disassembly of the spiral by pulling its end. (<b>b</b>) Representative structure of exosomes with cargos. Note that placenta alkaline phosphatase (PLAP) is a specific marker of placenta-derived EVs. ARFs, ADP ribosylation factors; CD, cluster of differentiation; ESCRT, endosomal sorting complex required for transport; LAMPs, lysosome-associated membrane glycoproteins; mRNA, messenger RNA; miRNA, microRNA; RABs, member of RAS superfamily of small G proteins; TfR, transferrin receptor.</p>
Full article ">Figure 3
<p>EV characterization methods for particle, protein, and miRNA evidence. ELISA: enzyme linked immunosorbent assay, EV: extracellular vesicle, LC-MS/MS: liquid chromatography-tandem mass spectrometry, miRNA: microRNA, PLAP: placental alkaline phosphatase protein, qRT-PCR: quantitative real-time polymerase chain reaction.</p>
Full article ">Figure 4
<p>Potential roles of placental debris and exosome changes during pregnancy-related complications. Placental debris and EVs (including exosomes and microvesicles) released from the placenta under different micro-environment conditions (such as hypoxia or high glucose) and their targeting of neighboring cells in the placenta and distant organs such as skeletal muscles. EVs are released by placental cells (such as syncytiotrophoblasts, cytotrophoblasts (CTs), and extravillous trophoblasts (EVTs)) and other cells in the placenta such as the placental mesenchymal stem cells. EVs from the placenta can enter maternal circulation and target distant cells such as skeletal muscles, endothelial cells, or vascular smooth muscle cells. Placenta-derived EVs’ concentration, content, and bioactivity changes in pregnancy and pregnancy-related disorders including GDM, PE, PTB, and FGR. GDM: gestational diabetes, PE: preeclampsia; FGR: fetal growth restriction, PTB: preterm birth.</p>
Full article ">Figure 5
<p>The possible roles of placenta-derived exosomes in the pathogenesis of Bart’s hydrops fetalis. Features of fetus with Bart’s hydrops are hepatosplenomegaly, cardiomegaly, ascites, pleural effusion, skin edema, and/or placentomegaly. Women with Bart’s hydropic fetuses can be presented with preeclampsia features called “mirror syndrome”. Placental hypoxia in Bart’s hydrops fetus is hypothesized to trigger the release of placental debris or placenta-derived exosomes which enter maternal circulation.</p>
Full article ">Figure 6
<p>An integrative diagnostic model combining current biomarkers and placenta-derived EVs for the identification of Bart’s hydrops fetalis throughout gestation. CTR: cardiothoracic diameter ratio, MCA-PSV: middle cerebral artery peak systolic velocity, MoM: multiple of median, MAFP: maternal alpha fetoprotein, uE3: unconjugated estriol.</p>
Full article ">
22 pages, 3944 KiB  
Article
Synthesis, Characterization and Biocompatibility Evaluation of Novel Chitosan Lipid Micro-Systems for Modified Release of Diclofenac Sodium
by Ana-Maria Raluca Pauna, Liliana Mititelu Tartau, Maria Bogdan, Andreea-Daniela Meca, Gratiela Eliza Popa, Ana Maria Pelin, Cristian Ilie Drochioi, Daniela Angelica Pricop and Liliana Lacramioara Pavel
Biomedicines 2023, 11(2), 453; https://doi.org/10.3390/biomedicines11020453 - 4 Feb 2023
Cited by 5 | Viewed by 2160
Abstract
The purpose of our study was the obtaining, characterization and biocompatibility estimation of novel carrier systems for diclofenac. Diclofenac is a potent nonsteroidal anti-inflammatory drug with frequent gastrointestinal side effects, impairing the quality of the patient’s life. Original diclofenac-loaded micro-vesicles coated with chitosan [...] Read more.
The purpose of our study was the obtaining, characterization and biocompatibility estimation of novel carrier systems for diclofenac. Diclofenac is a potent nonsteroidal anti-inflammatory drug with frequent gastrointestinal side effects, impairing the quality of the patient’s life. Original diclofenac-loaded micro-vesicles coated with chitosan were prepared and physico-chemical analyzed. We investigated their in vitro hemocompatibility and in vivo biocompatibility in rats. The animals were treated orally as follows: group 1 (Control): distilled water 0.3 mL/100 g body weight; Group 2 (CHIT): 0.3 mL/100 g body weight 0.5% chitosan solution; Group 3 (DCF): 15 mg/kg body weight diclofenac; Group 4 (DCF-ves): lipid vesicles loaded with diclofenac 15 mg/kg body weight. Blood samples were collected for assessing: red blood cells, hemoglobin, hematocrit and leukocyte formula. A series of specific parameters of the liver and kidney function, some markers of immune defense, as well as the activity of some enzymes involved in oxidative processes, were also investigated. At the end of the experiment, the animals were sacrificed and fragments of liver, kidney and stomach were collected for histopathological examination. No blood hemolysis was evidenced by the in vitro test with the administration of diclofenac vesicles. The animals treated with diclofenac lipid vesicles stabilized with chitosan did not display any notable differences in their hematological and biochemical profile compared to control animals. These data correlated with the histological results, which showed the absence of architectural changes in the examined tissues. Biological in vitro and in vivo evaluation revealed that the microvesicles containing diclofenac are biocompatible, with potential to be used as delivery systems to modify the drug release, thus making them an attractive candidate for biomedical applications. Full article
(This article belongs to the Section Drug Discovery, Development and Delivery)
Show Figures

Figure 1

Figure 1
<p>The procedure for obtaining chitosan-stabilized lipid vesicles with DCF.</p>
Full article ">Figure 2
<p>DIC (20×) optical microscopy images of chitosan-free DCF-ves (<b>a</b>) and chitosan-stabilized and dialyzed DCF-ves (<b>b</b>).</p>
Full article ">Figure 3
<p>Size distribution of DCF-ves.</p>
Full article ">Figure 4
<p>The dimensional histogram of DCF-ves coated with chitosan.</p>
Full article ">Figure 5
<p>Distribution of Zeta potential for DCF-vesicles.</p>
Full article ">Figure 6
<p>UV–vis Spectra for DCF in suspension (<b>a</b>) and calibration curve (<b>b</b>).</p>
Full article ">Figure 7
<p>The absorption spectra of DCF and DCF-ves (legend: a.u.–absorption units, λ–wavelength).</p>
Full article ">Figure 8
<p>The release kinetics of DCF from DCF-ves, respectively, from the DCF solution by the permeation method.</p>
Full article ">Figure 9
<p>Histopathological examination of liver fragments taken from rats treated with bi-distilled water (<b>a</b>), CHIT (<b>b</b>), DCF (<b>c</b>), DCF-ves (<b>d</b>)–Masson trichrome staining × 20.</p>
Full article ">Figure 10
<p>Histopathological examination of kidney fragments taken from rats treated with bi-distilled water (<b>a</b>), CHIT (<b>b</b>), DCF (<b>c</b>), DCF-ves (<b>d</b>)–Masson trichrome staining × 20.</p>
Full article ">Figure 11
<p>Histopathological examination of stomach fragments taken from rats treated with bi-distilled water (<b>a</b>), CHIT (<b>b</b>), DCF (<b>c</b>), DCF-ves (<b>d</b>)–Masson trichrome staining × 20.</p>
Full article ">
15 pages, 1292 KiB  
Article
The Association of Glucose Control with Circulating Levels of Red Blood Cell-Derived Vesicles in Type 2 Diabetes Mellitus Patients with Atrial Fibrillation
by Alexander A. Berezin, Zeljko Obradovic, Kristen Kopp, Tetiana A. Berezina, Michael Lichtenauer, Bernhard Wernly and Alexander E. Berezin
Int. J. Mol. Sci. 2023, 24(1), 729; https://doi.org/10.3390/ijms24010729 - 31 Dec 2022
Cited by 1 | Viewed by 2479
Abstract
Hyperglycemia is a trigger for structural alteration of red blood cells (RBCs) and their ability to release extracellular vesicles (EVs). The aim of the study was to elucidate whether glucose control in T2DM patients with concomitant HF and AF affects a circulating number [...] Read more.
Hyperglycemia is a trigger for structural alteration of red blood cells (RBCs) and their ability to release extracellular vesicles (EVs). The aim of the study was to elucidate whether glucose control in T2DM patients with concomitant HF and AF affects a circulating number of RBC-derived EVs. We prospectively included 417 T2DM patients with HF, 51 of them had atrial fibrillation and 25 healthy volunteers and 30 T2DM non-HF individuals. Clinical assessment, echocardiography examination and biomarker measures were performed at the baseline of the study. RBC-derived EVs were determined as CD235a+ PS+ particles by flow cytometry. NT-proBNP levels were measured by ELISA. AF patients with glycosylated hemoglobin (HbA1c) < 6.9% had lower levels of CD235a+ PS+ RBC-derived vesicles than those with HbA1c ≥ 7.0%. There were no significant differences in number of CD235a+ PS+ RBC-derived vesicles between patients in entire cohort and in non-AF sub-cohort with HbA1c < 6.9% and HbA1c ≥ 7.0%, respectively. Multivariate linear regression yielded that CD235a+ PS+ RBC-derived vesicles ≥ 545 particles in µL (OR = 1.06; 95% CI = 1.01–1.11, p = 0.044) independently predicted HbA1c ≥ 7.0%. Elevated levels of CD235a+ PS+ RBC-derived EVs independently predicted poor glycaemia control in T2DM patients with HF and AF. Full article
(This article belongs to the Special Issue Roles of Erythrocytes in Human Health and Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Amount of circulating CD235a+ PS+ RBC-derived vesicles: (<b>Left panel</b>) depicts results of flow cytometry measure of PS+ CD235a+ RBS-derived vesicles; Distinguishing populations of RBC-derived vesicles were determined at the first step of gaiting depending on their size (&lt;0.5 μm) using forward scatter and side scatter dot plot (<b>A</b>). Next, CD235a+ RBC- derived vesicles and PS+ RBC- derived vesicles were determined as double-positive events for labels indicated as CD235a+ and PS+ in T2DM non-HF (<b>B</b>), T2DM patients with HF (<b>C</b>) when compared with healthy volunteers (<b>D</b>). (<b>Right panel</b>) illustrate comparable proportion of circulating amount of CD235a+ PS+ RBC-derived vesicles in different cohorts of eligible individuals. Abbreviations: T2DM, type 2 diabetes mellitus; HF, heart failure; AF, atrial fibrillation, RBC, red blood cells; PS, phosphatidylserine.</p>
Full article ">Figure 2
<p>The amount of CD235a+ PS+ RBC-derived vesicles in T2DM patients with HF depending of glycemia control. Abbreviations: T2DM, type 2 diabetes mellitus; HF, heart failure; AF, atrial fibrillation, RBC, red blood cells; PS, phosphatidylserine; HbA1c, glycosilated hemoglobin.</p>
Full article ">Figure 3
<p>The predictive ability of circulating amount of CD235a+ PS+ RBC-derived vesicles for poor glycaemia control The Receive Operation Characteristics curve analysis. Abbreviations: AUC, area under curve; CI, confidence interval; RBC, red blood cells; PS, phosphatidylserine.</p>
Full article ">Figure 4
<p>Study design and patients’ procedure flow chart. Abbreviations: AF, atrial fibrillation; CABG, coronary artery bypass grafting; CV, cardiovascular; CKD, chronic kidney disease; GFR, glomerular filtration rate; HF, heart failure; HbA1c, glycated hemoglobin; HOMA-IR, Homeostatic Assessment Model of Insulin Resistance; N-terminal brain natriuretic pro-peptide; T2DM, type 2 diabetes mellitus; TIA, transient ischemic attack.</p>
Full article ">
18 pages, 4918 KiB  
Article
Nano-Sized Extracellular Vesicles Secreted from GATA-4 Modified Mesenchymal Stem Cells Promote Angiogenesis by Delivering Let-7 miRNAs
by Min Gong, Min Wang, Jie Xu, Bin Yu, Yi-Gang Wang, Min Liu, Muhammad Ashraf and Meifeng Xu
Cells 2022, 11(9), 1573; https://doi.org/10.3390/cells11091573 - 7 May 2022
Cited by 10 | Viewed by 2854
Abstract
We demonstrated previously that extracellular vesicles (EVs) released from mesenchymal stem cells (MSCs) play a critical role in angiogenesis. Here, we examine whether this pro-angiogenic efficacy is enhanced in EVs derived from MSCs overexpressing GATA-4 (MSCGATA−4). Methods and Results. EVs were [...] Read more.
We demonstrated previously that extracellular vesicles (EVs) released from mesenchymal stem cells (MSCs) play a critical role in angiogenesis. Here, we examine whether this pro-angiogenic efficacy is enhanced in EVs derived from MSCs overexpressing GATA-4 (MSCGATA−4). Methods and Results. EVs were isolated from MSCGATA-4 (EVGATA-4) and control MSCs transduced with an empty vector (EVnull). EVs from both cell types were of the same size and displayed similar molecular markers. Compared with EVnull, EVGATA-4 increased both a tube-like structure formation and spheroid-based sprouting of human umbilical vein endothelial cells (HUVECs). The EVGATA-4 increased the numbers of CD31-positive cells and hemoglobin content inside Matrigel plugs subcutaneously transplanted into mice for 2 weeks. Moreover, EVGATA-4 encapsulated higher levels of let-7 family miRs compared to EVnull. The transfer of exosomal let-7 miRs into HUVECs was recorded with an accompanied down-regulation of thrombospondin-1 (THBS1) expression, a major endogenous angiogenesis inhibitor. The loss-and-gain of function studies of let-7 miRs showed that let-7f knockdown significantly decreased EVGATA-4-mediated vascularization inside Matrigel plugs. In contrast, let-7f overexpression promoted HUVEC migration and tube formation. Conclusion. Our results indicate that EVs derived from genetically modified MSCs with GATA-4 overexpression had increased pro-angiogenic capacity due to the delivery of let-7 miRs that targeted THBS1 in endothelial cells. Full article
(This article belongs to the Special Issue Mesenchymal Stem Cell-Derived Extracellular Vesicles)
Show Figures

Figure 1

Figure 1
<p>A schematic diagram of the EV collection from cultured MSCs.</p>
Full article ">Figure 2
<p>MSC<sup>GATA−4</sup> construction and EVs identification. (<b>A</b>) Immunofluorescence staining showed GATA-4 overexpression in MSC<sup>GATA−4</sup> nucleus. (<b>B</b>) The mRNA of GATA-4 in MSCs determined by qRT-PCR, * <span class="html-italic">p</span> &lt; 0.05 vs. MSC<sup>null</sup>. (<b>C</b>) The morphology of EVs under transmission electron microscopy. (<b>D</b>) The size of EV<sup>null</sup> and EV<sup>GATA−4</sup> was measured using a Zetasizer Nano instrument. (<b>E</b>) Western blotting of CD9, CD63, and HSP70 in MSCs and EVs. (<b>F</b>) Western blotting of Calnexin in MSCs and EVs.</p>
Full article ">Figure 3
<p>EV<sup>GATA−4</sup> promoted tube-like structure formation and spheroid sprouting. (<b>A</b>) EVs mediated the tube-like structure formation in a concentration-related manner. (<b>B</b>) Representative images of capillary-like tube formation and quantitative analysis of the total tube length. (<b>C</b>) Representative images of HUVEC spheroids sprouting and quantitative analysis of the cumulative sprout length per spheroid. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. CON (BSA); * <span class="html-italic">p</span> &lt; 0.05 vs. EV<sup>null</sup>.</p>
Full article ">Figure 4
<p>EV<sup>GATA−4</sup> promoted angiogenesis in vivo. (<b>A</b>) The representative gross appearance of Matrigel™ plugs, which were implanted subcutaneously in mice for 14 days. (<b>B</b>) Hemoglobin content in the Matrigel™ plugs. (<b>C</b>) Immunofluorescence staining of CD31 in the sections of Matrigel™ plugs and quantification of the CD31-positive cells. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. CON (BSA); * <span class="html-italic">p</span> &lt; 0.05 vs. EV<sup>null</sup>.</p>
Full article ">Figure 5
<p>EVs mediated the transfer of let-7 miRs. (<b>A</b>) The up-regulation of let-7a, let-7d, let-7e, and let-7f in EV<sup>GATA-4</sup> was evaluated by qRT-PCR. (<b>B</b>) The quantitative analysis of let-7 miRs in HUVECs treated with EV<sup>null</sup> and EV<sup>GATA-4</sup>. (<b>C</b>) The HUVECs showed strong positivity for CD63 after culture with EV<sup>CD63-GFP</sup>. (<b>D</b>) Let-7f (red) was strongly expressed in HUVECs cultured with EV<sup>CD63-GFP/CY3-let-7f</sup>. * <span class="html-italic">p</span> &lt; 0.05 vs. EV<sup>null</sup>.</p>
Full article ">Figure 6
<p>EV<sup>GATA-4</sup>-mediated angiogenesis was related to the expression of let-7. (<b>A</b>) The expression of let-7f in MSC<sup>G4 + anti-let-7f</sup> and EV<sup>G4 + anti-let-7f</sup>, as well as their control. (<b>B</b>) The expression of let-7f in HUVECs treated with EV<sup>G4 + anti-let-7f</sup> and EV<sup>G4-Scr</sup>. (<b>C</b>) Representative images of capillary-like tube formation and quantitative analysis of the total tube length following EV treatment. (<b>D</b>) Immunofluorescence staining of CD31 in the sections of Matrigel™ plugs, and the quantification of the CD31-positive cells. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. MSC<sup>G4 + Scr</sup>; <b>*</b> <span class="html-italic">p</span> &lt; 0.05 vs. EV<sup>G4 + Scr</sup>. (EV<sup>G4 + Scr</sup> = EV<sup>GATA-4 + scrambled</sup>; EV<sup>G4 + anti-let-7f</sup> = EV<sup>GATA-4 + anti-let-7f</sup>).</p>
Full article ">Figure 7
<p>Exosomal let-7 miRs functionally down-regulated the target gene THBS1 in HUVECs. (<b>A</b>) An example of complementarity between let-7 miRs and the THBS1 3′UTR. (<b>B</b>,<b>C</b>) Quantitative data for dual-luciferase reporter assay in HUVECs treated with EV<sup>null</sup> or EV<sup>GATA-4</sup>. (<b>D</b>) The THBS1 protein levels in HUVECs following different treatments determined by Western blotting. (<b>E</b>) The protein level of THBS1 in HUVECs treated with EVs. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. CON (BSA); <b>*</b> <span class="html-italic">p</span> &lt; 0.05 vs. EV<sup>null</sup>; and <sup>@</sup> <span class="html-italic">p</span> &lt; 0.05 vs. EV<sup>G4-Scr</sup>, respectively. (EV<sup>G4 + Scr</sup> = EV<sup>GATA-4 + Scrambled</sup>; EV<sup>G4 + anti-let-7f</sup> = EV<sup>GATA-4 + anti-let-7f</sup>).</p>
Full article ">Figure 8
<p>Let-7f overexpression in HUVECs promoted tube-like structure formation and migration. (<b>A</b>) Representative images of HUVECs with let-7f overexpression (HUVEC<sup>let-7f</sup>), and their negative control (HUVEC<sup>Scrambled</sup>). (<b>B</b>) The expression of let-7f in HUVECs. (<b>C</b>) The THBS1 protein in HUVECs transfected with let-7f. (<b>D</b>) Representative images of capillary-like tube formation and quantitative analysis of the total tube length. (<b>E</b>) Representative images of HUVEC migration and quantitative analysis. * <span class="html-italic">p</span> &lt; 0.05 vs. HUVEC<sup>Scr</sup>. (HScr = HUVEC<sup>Scrambled</sup>, H<sup>let-7f</sup> = HUVEC<sup>let-7f</sup>.)</p>
Full article ">
19 pages, 1382 KiB  
Article
MicroRNAs miR-451a and Let-7i-5p Profiles in Circulating Exosomes Vary among Individuals with Different Sickle Hemoglobin Genotypes and Malaria
by Keri Oxendine Harp, Alaijah Bashi, Felix Botchway, Yvonne Dei-Adomakoh, Shareen A. Iqbal, Michael D. Wilson, Andrew A. Adjei, Jonathan K. Stiles and Adel Driss
J. Clin. Med. 2022, 11(3), 500; https://doi.org/10.3390/jcm11030500 - 19 Jan 2022
Cited by 9 | Viewed by 3023
Abstract
Sickle cell disease (SCD) occurs when two alleles of mutated hemoglobin (HbS or HbC) are inherited (HbSS and HbSC) rather than one (HbAS or HbAC), which indicates a person carries the sickle cell trait. The high prevalence of these two alleles in Africa [...] Read more.
Sickle cell disease (SCD) occurs when two alleles of mutated hemoglobin (HbS or HbC) are inherited (HbSS and HbSC) rather than one (HbAS or HbAC), which indicates a person carries the sickle cell trait. The high prevalence of these two alleles in Africa have been associated with reduced malaria susceptibility. Recent in vitro research has been shown that microRNAs (miRNAs) miR-451a and let-7i-5p are differentially expressed in HbSS erythrocytes compared to healthy controls (HbAA) and are overexpressed in Plasmodium-infected malaria erythrocytes. However, these miRNAs have not been fully examined in the plasma of people with different sickle hemoglobin genotypes. Plasma circulating miRNAs are commonly encapsulated in extracellular vesicles, such as exosomes, and are thought to play a role in disease development. Circulating exosomal miR-451a and let-7i-5p were quantified from individuals with various hemoglobin genotypes (HbAA, HbAS, HbAC, HbSS, HbSC, and HbCC) with (+) and without (−) malaria. The results showed a higher level of exosomal let-7i-5p and miR-451a in HbSS-. Exosomal let-7i-5p and miR-451a levels were lower in HbSS+ compared to other genotypes. Based on the area under the curve (AUC) of the Receiver Operating Characteristics (ROCs), both exosomal miRNAs may be useful disease biomarkers for SCD with malaria. Finally, miR-451a and let-7i-5p modulate genes involved in inflammation, making them potential biomarkers of pathogenesis for both diseases. Full article
(This article belongs to the Section Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>RT-qPCR results for exosomal miR-451a and let-7i-5p expressed as log of fold change (ΔΔCt) in a bar graph expressing mean and standard deviation. ΔΔCt was created using RNU6 as endogenous control and HbAA− as the control group in Excel using the Livak method [<a href="#B28-jcm-11-00500" class="html-bibr">28</a>] (<b>A</b>) There was a significant difference in relative expression of exosomal let-7i-5p for HbSS− vs. HbAC− (<span class="html-italic">p</span> = 0.006), HbSS− vs. HbCC− (<span class="html-italic">p</span> = 0.04), and HbSS− vs. HbSC− (<span class="html-italic">p</span> = 0.004). (<b>B</b>) There was a significant difference for expression of exosomal miR-451a between HbAA− vs. HbCC− (<span class="html-italic">p</span> &lt; 0.0001), HbAA− vs. HbSC− (<span class="html-italic">p</span> = 0.0004), HbAS− vs. HbCC− (<span class="html-italic">p</span> = 0.0004), HbAS− vs. HbSC− (<span class="html-italic">p</span> = 0.003), HbCC− vs. HbSS− (<span class="html-italic">p</span> = 0.0003), and HbSC− vs. HbSS− (<span class="html-italic">p</span> = 0.003). (<b>C</b>) There was a significant difference for exosomal let-7i-5p between HbAA− vs. HbSS+ (<span class="html-italic">p</span> = 0.002), HbAA+ vs. HbSS+ (<span class="html-italic">p</span> = 0.0008), HbAC+ vs. HbSS+ (<span class="html-italic">p</span> = 0.01), and HbAS+ vs. HbSS+ (<span class="html-italic">p</span> = 0.01). (<b>D</b>) There was a significant difference for exosomal miR-451a between HbSS+ vs. HbAA− (<span class="html-italic">p</span> &lt; 0.0001), HbSS+ vs. HbAA+ (<span class="html-italic">p</span> &lt; 0.0001), HbSS+ vs. HbAS+ (<span class="html-italic">p</span> &lt; 0.0001), HbSC+ vs. HbSS+ (<span class="html-italic">p</span> = 0.04), and HbSS+ vs. HbAC+ (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 2
<p>Correlation of exosomal let-7i-5p and miR-451a expression levels among all genotypes. Two-tailed Pearson correlation with 95% confidence bands of let-7i-5p and miR-451a between genotypes. (<b>A</b>) For HbAA−, HbAS−, HbSS−, HbAC−, HbSC−, and HbCC−, R<sup>2</sup> = 0.45 and <span class="html-italic">p</span> &lt; 0.0001. (<b>B</b>) For HbAA+, HbAS+, HbSS+, HbAC+, and HbSC+, R2 = 0.76 and <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Pearson correlation on each miRNA versus complete blood counts (CBCs) (RBC, WBC, Hb, and PLT) for each genotype with and without malaria. RBCs = red blood cells; WBCs = white blood cells; Hb = hemoglobin; PLT = platelet. Only 7 correlations were significant and shown. (<b>A</b>) For HbAA+, correlation between exosomal let-7i-5p and RBC resulted in R<sup>2</sup> = 0.23 and <span class="html-italic">p</span> = 0.01. (<b>B</b>) For HbSS+, correlation between exosomal let-7i-5p and Hb was R<sup>2</sup> = 0.4 and <span class="html-italic">p</span> = 0.04. (<b>C</b>) For HbSS+, correlation between exosomal miR-451a and Hb was R<sup>2</sup> = 0.42 and <span class="html-italic">p</span> = 0.04. (<b>D</b>) For HbSS+, correlation between exosomal let-7i-5p and PLT was R<sup>2</sup> = 0.4 and <span class="html-italic">p</span> = 0.04. (<b>E</b>) For HbSC−, correlation between exosomal miR-451a and Hb resulted in R<sup>2</sup> = 0.36 and <span class="html-italic">p</span> = 0.02. (<b>F</b>) For HbSC−, correlation between exosomal let-7i-5p and PLT resulted in R<sup>2</sup> = 0.31 and <span class="html-italic">p</span> = 0.03. (<b>G</b>) For HbSC−, correlation between exosomal miR-451a and PLT resulted in R<sup>2</sup> = 0.3 and <span class="html-italic">p</span> = 0.03.</p>
Full article ">Figure 4
<p>ROC analysis for exosomal miR-451a and let-7i-5p (<b>A</b>) miR-451a, SCA+ (HbSS+ and HbSC+ combined) vs. HbAA+, AUC = 0.8365, <span class="html-italic">p</span> = 0.0003; (<b>B</b>) let-7i-5p, SCA+ vs. HbAA+, AUC = 0.8280, <span class="html-italic">p</span> = 0.0002; (<b>C</b>) miR-451a, SCA+ vs. HbAA−, AUC = 0.8457, <span class="html-italic">p</span> = 0.0001; (<b>D</b>) let-7i-5p, SCA+ vs. HbAA−, AUC = 0.8238, <span class="html-italic">p</span> = 0.0001; (<b>E</b>) miR-451a, SCA+ vs. SCT+ (HbAS+ and HbAC+ combined), AUC = 0.8250, <span class="html-italic">p</span> = 0.0009; (<b>F</b>) let-7i-5p, SCA+ vs. SCT+, AUC = 0.8222, <span class="html-italic">p</span> = 0.0007; (<b>G</b>) miR-451a, SCA+ vs. SCT− (HbAS− and HbAC− combined), AUC = 0.7604, <span class="html-italic">p</span> = 0.0023; (<b>H</b>) let-7i-5p, SCA+ vs. SCT−, AUC = 0.7407, <span class="html-italic">p</span> = 0.0033.</p>
Full article ">
10 pages, 2897 KiB  
Article
Liposomal Artificial Red Blood Cell-Based Carbon Monoxide Donor Is a Potent Renoprotectant against Cisplatin-Induced Acute Kidney Injury
by Kazuaki Taguchi, Yuto Suzuki, Moeko Tsutsuura, Kana Hiraoka, Yuki Watabe, Yuki Enoki, Masaki Otagiri, Hiromi Sakai and Kazuaki Matsumoto
Pharmaceutics 2022, 14(1), 57; https://doi.org/10.3390/pharmaceutics14010057 - 27 Dec 2021
Cited by 6 | Viewed by 2956
Abstract
Cisplatin (CDDP) is an essential anti-tumor agent for chemotherapeutic regimens against various types of cancer. However, the progression of nephrotoxicity, which is the main adverse effect of CDDP, leads to discontinuation of CDDP chemotherapy. Therefore, development of a renoprotectant against CDDP-induced nephrotoxicity is [...] Read more.
Cisplatin (CDDP) is an essential anti-tumor agent for chemotherapeutic regimens against various types of cancer. However, the progression of nephrotoxicity, which is the main adverse effect of CDDP, leads to discontinuation of CDDP chemotherapy. Therefore, development of a renoprotectant against CDDP-induced nephrotoxicity is crucial. Here, the potential of a carbon monoxide (CO)-loaded hemoglobin-vesicle (CO-HbV) as a renoprotectant for CDDP-induced nephrotoxicity was evaluated for its renoprotective effects against CDDP-induced nephrotoxicity, inhibitory effects on the anti-tumor activity of CDDP, and anti-tumor activity. In healthy mice, after pretreatment with either saline, HbV, or CO-HbV prior to CDDP administration, only the CO-HbV pretreatment group ameliorated the progression of CDDP-induced nephrotoxicity by suppressing apoptosis via caspase-3. In experiments using B16-F10 melanoma cells, the half-maximal inhibitory concentration of CDDP decreased with co-incubation with CO-HbV, owing to the anti-tumor activity of CO. CO-HbV pretreatment had no impact on the anti-tumor activity of CDDP in B16-F10 melanoma cell-bearing mice, which was consistent with the results of the cell experiment. Furthermore, CO-HbV pretreatment improved body growth and survival rates. In conclusion, CO-HbV pretreatment is a potent renoprotectant for CDDP-induced nephrotoxicity, allowing treatment with CDDP to be conducted without failure of cancer treatment. Full article
(This article belongs to the Collection Advanced Pharmaceutical Science and Technology in Japan)
Show Figures

Figure 1

Figure 1
<p>Biochemical analysis of kidney injury. The levels of (<b>A</b>) blood urea nitrogen (BUN) and (<b>B</b>) creatinine at 96 h after each sample treatment in cisplatin (CDDP) mice model. The data represent the mean ± standard deviation (S.D.). Control (<span class="html-italic">n</span> = 8), saline (<span class="html-italic">n</span> = 7), Hemoglobin-vesicle (HbV) (<span class="html-italic">n</span> = 8), carbon monoxide (CO)-loaded HbV (CO-HbV) (<span class="html-italic">n</span> = 8); ** <span class="html-italic">p</span> &lt; 0.01 vs. control, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 vs. Saline, † <span class="html-italic">p</span> &lt; 0.05, †† <span class="html-italic">p</span> &lt; 0.01 vs. HbV.</p>
Full article ">Figure 2
<p>Representative imaging of kidney stained by PAS at 96 h after each sample treatment in cisplatin (CDDP) mouse model. (<b>A</b>) Control mice (without CDDP treatment), CDDP mouse model treated with (<b>B</b>) saline, (<b>C</b>) Hemoglobin-vesicle (HbV), and (<b>D</b>) carbon monoxide (CO)-loaded HbV (CO-HbV). Scale bar: 100 µm.</p>
Full article ">Figure 3
<p>Representative TUNEL image of kidney at 96 h after each sample treatment in cisplatin (CDDP) mouse model. (<b>A</b>) Control mice (without CDDP treatment), CDDP mouse model treated with (<b>B</b>) saline, (<b>C</b>) Hemoglobin-vesicle (HbV), and (<b>D</b>) carbon monoxide (CO)-loaded HbV (CO-HbV). The images shown as merged images of TUNEL-positive cells (green) and DAPI staining (blue) (200× magnification). (<b>E</b>) Quantitative image analysis of TUNEL (green) and DAPI (blue) positive cells (white arrows). The data represents the mean ± S.D. Control (<span class="html-italic">n</span> = 8), Saline (<span class="html-italic">n</span> = 7), HbV (<span class="html-italic">n</span> = 8), CO-HbV (<span class="html-italic">n</span> = 8); # <span class="html-italic">p</span> &lt; 0.05 vs. Saline.</p>
Full article ">Figure 4
<p>The effect of each treatment on the caspase-3 activation in kidney of cisplatin (CDDP) mouse model. (<b>A</b>) Representative protein expression and (<b>B</b>) quantitative analysis of band intensity of caspase-3 cleavage protein in kidney at 96 h after sample treatment. The data represents the mean ± S.D. Control (<span class="html-italic">n</span> = 8), Saline (<span class="html-italic">n</span> = 7), Hemoglobin-vesicle (HbV) (<span class="html-italic">n</span> = 8), carbon monoxide (CO)-loaded HbV (CO-HbV) (<span class="html-italic">n</span> = 8); ## <span class="html-italic">p</span> &lt; 0.01 vs. Saline. P; positive control.</p>
Full article ">Figure 5
<p>The effects of carbon monoxide (CO)-loaded Hemoglobin-vesicle (CO-HbV) on tumor cells. (<b>A</b>) Dose-dependent cytotoxicity effect of CO-HbV on B16-F10 melanoma cells. (<b>B</b>) The inhibitory effects of CO-HbV on anti-tumor activity of cisplatin (CDDP) toward B16-F10 melanoma cells. The data represent the mean ± S.D. (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 6
<p>Effect of carbon monoxide (CO)-loaded Hemoglobin-vesicle (CO-HbV) on the anti-tumor efficacy of the conventional cisplatin (CDDP) treatment in B16-F10 bearing mice. The profiles of (<b>A</b>) body weight, (<b>B</b>) survival rate and (<b>C</b>) tumor volume until 14 days after the commencement of CDDP administrations. The data represent the mean ± S.D. (<span class="html-italic">n</span> = 10). ** <span class="html-italic">p</span> &lt; 0.01 vs. Control, ## <span class="html-italic">p</span> &lt; 0.01 vs. Saline, † <span class="html-italic">p</span> &lt; 0.05 vs. HbV. The body weight differences of saline, HbV, and CO-HbV groups were significant (<span class="html-italic">p</span> &lt; 0.01) from day 2 to day 10, from day 2 to day 11, and from day 2 to day 14, respectively, compared to the control group. The tumor volume of saline, HbV and CO-HbV group were significantly different (<span class="html-italic">p</span> &lt; 0.01) from day 6 to day 10, from day 6 to day 11, and from day 6 to day 14, respectively, compared to the control group.</p>
Full article ">
28 pages, 1815 KiB  
Review
The Role of RBC Oxidative Stress in Sickle Cell Disease: From the Molecular Basis to Pathologic Implications
by Qinhong Wang and Rahima Zennadi
Antioxidants 2021, 10(10), 1608; https://doi.org/10.3390/antiox10101608 - 13 Oct 2021
Cited by 33 | Viewed by 5850
Abstract
Sickle cell disease (SCD) is an inherited monogenic disorder and the most common severe hemoglobinopathy in the world. SCD is characterized by a point mutation in the β-globin gene, which results in hemoglobin (Hb) S production, leading to a variety of mechanistic and [...] Read more.
Sickle cell disease (SCD) is an inherited monogenic disorder and the most common severe hemoglobinopathy in the world. SCD is characterized by a point mutation in the β-globin gene, which results in hemoglobin (Hb) S production, leading to a variety of mechanistic and phenotypic changes within the sickle red blood cell (RBC). In SCD, the sickle RBCs are the root cause of the disease and they are a primary source of oxidative stress since sickle RBC redox state is compromised due to an imbalance between prooxidants and antioxidants. This imbalance in redox state is a result of a continuous production of reactive oxygen species (ROS) within the sickle RBC caused by the constant endogenous Hb autoxidation and NADPH oxidase activation, as well as by a deficiency in the antioxidant defense system. Accumulation of non-neutralized ROS within the sickle RBCs affects RBC membrane structure and function, leading to membrane integrity deficiency, low deformability, phosphatidylserine exposure, and release of micro-vesicles. These oxidative stress-associated RBC phenotypic modifications consequently evoke a myriad of physiological changes involved in multi-system manifestations. Thus, RBC oxidative stress in SCD can ultimately instigate major processes involved in organ damage. The critical role of the sickle RBC ROS production and its regulation in SCD pathophysiology are discussed here. Full article
Show Figures

Figure 1

Figure 1
<p>Generation of reactive oxygen species. Oxygen readily accepts free electrons produced by normal oxidative metabolism, generating the unstable superoxide anions (O<sub>2</sub><sup>•−</sup>), which are scavenged by superoxide dismutase (SOD) and transformed into hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>) or rapidly converted into peroxynitrite (ONOO<sup>−</sup>) when reacting with bioactive nitric oxide (<sup>•</sup>NO). The H<sub>2</sub>O<sub>2</sub> converts to water by catalase. In the Harber–Weiss reaction, hydroxyl radicals (<sup>•</sup>OH) are generated in the presence of H<sub>2</sub>O<sub>2</sub> and iron ions.</p>
Full article ">Figure 2
<p>The role of sickle RBCs in vaso-occlusion. Sickle RBCs interact with and activate the vascular endothelium, leukocytes, and platelets, and bind to coagulation factors and plasma proteins forming aggregates. These adhesive interactions progressively induce the occlusion of blood vessels and blood stasis.</p>
Full article ">Figure 3
<p>Potential contributions of sickle RBCs to venous thrombosis in SCD. Increased ROS levels in sickle RBCs impair membrane structure and function, leading to membrane integrity loss and reduced deformability, thus negatively modulating sickle RBC quality. This leads to a reduced flow of sickle RBC through the microcirculation. Venous thrombi form slowly in the presence of low flow or stasis and they are rich in both RBCs and fibrin. In the vasculature, sickle RBCs aggregate into stacked rouleaux structures, increasing blood viscosity. These sickle RBCs also adhere to the endothelium and participate in thrombin generation within thrombi. Once incorporated into the venous thrombus, sickle RBCs increase clot size and reduce both clot permeability and susceptibility to lysis. In SCD, sickle RBCs and sickle RBC-derived micro-particles also adhere to the vessel wall or extracellular matrix; activate endothelial cells, platelets, and other blood cells; and enhance local thrombin generation during thrombosis.</p>
Full article ">Figure 4
<p>Activation of the coagulation cascade by sickle RBC-derived microparticles and tissue factors. Phosphatidylserine-positive microparticles (PS-positive MPs) derived from sickle RBCs and sickle RBC-free hemoglobin-induced tissue factor (TF) release enhances thrombosis in SCD by activating the coagulation cascade.</p>
Full article ">Figure 5
<p>Activation of the complement system by sickle RBC-derived micro-vesicles in SCD. Micro-vesicles (MVs) derived from sickle RBCs can initiate activation of the complement system by cleaving C3 and C5 with C3 convertase and C5 convertase, respectively, leading to the release of the anaphylatoxins C3a and C5a, respectively.</p>
Full article ">
23 pages, 6011 KiB  
Article
Proteomic Analysis of Estrogen-Mediated Enhancement of Mesenchymal Stem Cell-Induced Angiogenesis In Vivo
by Maria Cristina Mihai, Mirel Adrian Popa, Viorel Iulian Șuică, Felicia Antohe, Edwin K. Jackson, Brigitte Leeners, Maya Simionescu and Raghvendra K. Dubey
Cells 2021, 10(9), 2181; https://doi.org/10.3390/cells10092181 - 24 Aug 2021
Cited by 4 | Viewed by 3155
Abstract
Therapeutic use of mesenchymal stem cells (MSCs) for tissue repair has great potential. MSCs from multiple sources, including those derived from human umbilical matrix, namely Wharton’s jelly, may serve as a resource for obtaining MSCs. However, low in vivo engraftment efficacy of MSCs [...] Read more.
Therapeutic use of mesenchymal stem cells (MSCs) for tissue repair has great potential. MSCs from multiple sources, including those derived from human umbilical matrix, namely Wharton’s jelly, may serve as a resource for obtaining MSCs. However, low in vivo engraftment efficacy of MSCs remains a challenging limitation. To improve clinical outcomes using MSCs, an in-depth understanding of the mechanisms and factors involved in successful engraftment is required. We recently demonstrated that 17β-estradiol (E2) improves MSCs in vitro proliferation, directed migration and engraftment in murine heart slices. Here, using a proteomics approach, we investigated the angiogenic potential of MSCs in vivo and the modulatory actions of E2 on mechanisms involved in tissue repair. Specifically, using a Matrigel® plug assay, we evaluated the effects of E2 on MSCs-induced angiogenesis in ovariectomized (OVX) mice. Moreover, using proteomics we investigated the potential pro-repair processes, pathways, and co-mechanisms possibly modified by the treatment of MSCs with E2. Using RT-qPCR, we evaluated mRNA expression of pro-angiogenic molecules, including endoglin, Tie-2, ANG, and VEGF. Hemoglobin levels, a marker for blood vessel formation, were increased in plugs treated with E2 + MSCs, suggesting increased capillary formation. This conclusion was confirmed by the histological analysis of capillary numbers in the Matrigel® plugs treated with E2 + MSC. The LC-MS screening of proteins obtained from the excised Matrigel® plugs revealed 71 proteins that were significantly altered following E2 exposure, 57 up-regulated proteins and 14 down-regulated proteins. A major result was the association of over 100 microRNA molecules (miRNAs) involved in cellular communication, vesicle transport, and metabolic and energy processes, and the high percentage of approximately 25% of genes involved in unknown biological processes. Together, these data provide evidence for increased angiogenesis by MSCs treated with the sex hormone E2. In conclusion, E2 treatment may increase the engraftment and repair potential of MSCs into tissue, and may promote MSC-induced angiogenesis after tissue injury. Full article
(This article belongs to the Section Cells of the Cardiovascular System)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>E2 enhances angiogenesis effects in Matrigel<sup>®</sup> plugs containing MSCs. (<b>A</b>) Exhibits a macroscopic view of Matrigel<sup>®</sup> plugs from each group—sham-operated or OVX—in presence or absence of E2 (100 nM): C57Bl mice injected with Matrigel<sup>®</sup> only, Matrigel<sup>®</sup> + cells (Matrigel<sup>®</sup> + MSC), or Matrigel<sup>®</sup> + hormone-treated cells (Matrigel<sup>®</sup> + MSC + E2). (<b>B</b>) Displays measures of hemoglobin content using Drabkin’s reagent kit of Matrigel<sup>®</sup> implants from both groups. The levels of this angiogenesis marker were amplified in MSCs stimulated with the sex hormone (MSC + E2 ± OVX) in comparison with control (MSCs only). Each value corresponds to mean ± SEM (<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 compared with control. (<b>C</b>) Descriptive photomicrographs of Matrigel<sup>®</sup>-derived blood capillaries/vessels embedded in paraffin and stained with hematoxylin &amp; eosin, and DAPI; Orange arrows indicate blood vessel structures; scale bar—100 μm. (<b>D</b>) Quantification of the new blood vessels was presented as mean ± SD (<span class="html-italic">n</span> = 2), ** <span class="html-italic">p</span> &lt; 0.01, and * <span class="html-italic">p</span> &lt; 0.001 compared with control. Note: E2, 17-beta estradiol; MSCs, Wharton’s jelly-derived mesenchymal stem cells; OVX, ovariectomy surgery.</p>
Full article ">Figure 2
<p>Liquid chromatography-mass spectrometry (LC-MS) testing revealed that E2 up-regulated proteins involved in angiogenesis and tissue remodeling. (<b>A</b>) represents the entire proteomic profile data of the in vivo Matrigel<sup>®</sup> plugs for each group (sham-operated and OVX) after LC-MS and bioinformatic analysis. A total of 2381 proteins were collectively identified in both control implants and Matrigel<sup>®</sup> plugs containing MSCs treated with E2. The relative quantitative analysis revealed 57 up-regulated and 14 down-regulated proteins (<span class="html-italic">p</span>-value &lt; 0.05). (<b>B</b>) illustrates statistical analysis (using Panther) of the protein pathways modulated by E2 presence in MSCs. Analyzing the proteins from excised Matrigel<sup>®</sup> plugs (containing E2 treated MSCs), we discovered proteins mainly involved in angiogenesis processes. VEGF (vascular endothelial growth factor), Ang-1 (angiopoietin 1), Tie-2 (angiopoietin receptor 2), and eNOS (endothelial nitric oxide synthase).</p>
Full article ">Figure 3
<p>Analysis of the differentially expressed proteins modulated by E2 shows a significant over-representation (False Discovery Rate corrected <span class="html-italic">p</span>-value &lt; 0.05) of the proteins participating in biological pathways, biological processes, cellular components and molecular functions. FunRich analysis of all the 2381 proteins which were commonly identified in both vehicle treated control MSCs and MSCs treated with E2 were characterized on their cellular components (<b>A</b>), biological pathways (<b>B</b>), molecular functions (<b>B</b>) and biological processes (<b>C</b>). DNA-BFTA, DNA-binding transcription factor activity; RNA-PII-S, RNA polymerase II-specific; and Clathrin-CEVM, Clathrin-coated endocytic vesicle membrane.</p>
Full article ">Figure 4
<p>FunRich (Uniprot database) analyze of the proteins differentially expressed by E2 stimulation shows a significant over-representation (False Discovery Rate corrected <span class="html-italic">p</span>-value &lt; 0.05) of the proteins participating in different cellular components and identified by Reactome pathways. The screening of the proteins involved in the paracrine function of the MSCs are represented as percentage (%) from the total of the cellular protein components.</p>
Full article ">Figure 5
<p>UniProt ID of the detected proteins reveals a total of 321 miRNAs through conversion by using FunRich miRNA enrichment tools. (<b>A</b>) shows miRNA enrichment based on UniProt ID protein’s conversion characterized on their biological process, (<b>B</b>) their molecular function, and (<b>C</b>) their cellular component.</p>
Full article ">Figure 6
<p>UniProt ID of the detected proteins reveal total of 321 miRNAs through conversion by using FunRich miRNA enrichment tools. Bar graph depicts miRNA enrichment based on UniProt ID protein’s conversion characterized on their Biological Pathway.</p>
Full article ">Figure 7
<p>Quantification of pro-angiogenic genes in Matrigel<sup>®</sup> plugs by RT-qPCR analysis shows enhanced levels of VEGFR-2, VEGF-A, Ang-1, Tie-2, and Endoglin modulated by E2 treatment of MSCs inserted in the Matrigel<sup>®</sup> plugs. Experiments were performed in triplicate and the results normalized to GAPDH. * <span class="html-italic">p</span> &lt; 0.05 versus Control (plugs with MSC only). Values represent the mean ± SEM from three separate experiments conducted in triplicates.</p>
Full article ">
13 pages, 1696 KiB  
Article
Large Platelet and Endothelial Extracellular Vesicles in Cord Blood of Preterm Newborns: Correlation with the Presence of Hemolysis
by Andrea Hujacova, Jan Sirc, Kristyna Pekarkova, Tereza Brozova, Marie Kostelanska, Jakub Soukup, Tibor Mosko, Karel Holada and Zbynek Stranak
Diagnostics 2021, 11(8), 1316; https://doi.org/10.3390/diagnostics11081316 - 22 Jul 2021
Cited by 2 | Viewed by 2129
Abstract
Different biomarkers are investigated to detect the causes of severe complications in preterm infants. Extracellular vesicles (EVs) are recognized as an important part of cell-to-cell communication, and their increased levels were reported in numerous pathological states. We aimed to increase our knowledge about [...] Read more.
Different biomarkers are investigated to detect the causes of severe complications in preterm infants. Extracellular vesicles (EVs) are recognized as an important part of cell-to-cell communication, and their increased levels were reported in numerous pathological states. We aimed to increase our knowledge about the incidence of platelet and endothelial EVs in cord blood of preterm newborns using conventional flow cytometry. The presence of platelet (CD36+CD41+), activated platelet (CD41+CD62+), and endothelial (CD31+CD105+) EVs was analyzed. Immune electron microscopy was used to confirm the presence of EVs and the specificity of their labeling. The size of detected extracellular vesicles was in the range 400–2000 nm. The differences in the counts of EVs between the preterm and control group were not significant and no correlation of EVs count with gestation age was recorded. Cord blood plasma samples with free hemoglobin level > 1 mg/mL had more than threefold higher counts of CD36+CD41+ and CD41+CD62+ EVs (p < 0.001), while the count of CD31+CD105+ EVs was only moderately increased (p < 0.05). Further studies utilizing cytometers with improved sensitivity are needed to confirm that the analysis of large platelet and endothelial EVs mirrors the quantitative situation of their whole plasma assemblage. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Flow cytometry analysis of large EVs in cord blood plasma. Standard silica and polystyrene (PS) beads were utilized to create EV gate in the SSC-H/FSC-H scattergram (<b>A</b>). Typical scattergram signal of cord blood plasma extracellular vesicles (<b>B</b>). Only events inside the gate were analyzed. Representative density plot of negative control sample (CTRL) labeled with isotype IgG1 control MAbs (<b>C</b>). Representative density plots of samples labeled with CD41 PE + CD36 FITC (<b>D</b>), CD41 FITC + CD62 PE (<b>E</b>), and CD31 FITC + CD105 PE (<b>F</b>) MAbs. Only double fluorescence positive events in the upper right quadrant were included in the analysis.</p>
Full article ">Figure 2
<p>Immune electron microscopy demonstrates the presence of platelet EVs in cord blood plasma. Samples were labeled with CD41 PE MAb and secondary antibody conjugated with colloidal gold followed by staining with uranyl acetate. Some of visualized EVs are decorated with colloidal gold ((<b>A</b>,<b>C</b>) black arrow) and some are non-labeled (<b>A</b>,<b>B</b>). The round hollow structures are artifacts present on carbon grids (arrowhead).</p>
Full article ">Figure 3
<p>Counts of cord blood plasma large EVs plotted against newborn gestational age. (<b>A</b>) Platelet (CD41+CD36+), (<b>B</b>) activated platelet (CD41+CD62+), and (<b>C</b>) endothelial (CD31+CD105+) EVs in preterm newborns (PRTM; circles, <span class="html-italic">n</span> = 20) and control newborns (CTRL; squares, <span class="html-italic">n</span> = 10). Gestational age (GA) is in weeks. Red symbols represent hemolytic samples (Hb &gt; 0.1 mg/mL).</p>
Full article ">Figure 4
<p>Counts of large EVs in hemolytic (H+) and non-hemolytic (H−) cord blood plasma samples. Platelet ((<b>A</b>) CD41+CD36+), activated platelet ((<b>B</b>) CD41+CD62+), and endothelial ((<b>C</b>) CD31+CD105+) EV counts in hemolytic (H+; squares, <span class="html-italic">n</span> = 7) and non-hemolytic (H−; circles, <span class="html-italic">n</span> = 23) samples. The medians are shown. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Comparison of median fluorescence intensity of labeled large EVs in cord blood plasma of PRTM and CTRL. Platelet (CD41+CD36+), activated platelet (CD41+CD62+), and endothelial (CD31+CD105+) EVs in preterm newborns (PRTM; circles, <span class="html-italic">n</span> = 20) and control newborns (CTRL; squares, <span class="html-italic">n</span> = 10) were analyzed. MFI of EVs labeled with CD41 PE (<b>A</b>), CD36 FITC (<b>B</b>), CD41 FITC (<b>C</b>), CD62 PE (<b>D</b>), CD31 FITC (<b>E</b>), and CD105 PE (<b>F</b>) are presented. Red symbols represent hemolytic samples (Hb &gt; 0.1 mg/mL). The medians are shown. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
17 pages, 5814 KiB  
Article
Centrifugation Removes a Population of Large Vesicles, or “Macroparticles,” Intermediate in Size to RBCs and Microvesicles
by Michael C. Larson, Neil Hogg and Cheryl A. Hillery
Int. J. Mol. Sci. 2021, 22(3), 1243; https://doi.org/10.3390/ijms22031243 - 27 Jan 2021
Cited by 3 | Viewed by 2289
Abstract
Microparticles or microvesicles (MPs/MVs) are sub-cellular vesicles with a growing number of known biological functions. Microvesicles from a variety of parent cells within the vascular system increase in numerous pathological states. Red blood cell-derived MVs (RMVs) are relatively less studied than other types [...] Read more.
Microparticles or microvesicles (MPs/MVs) are sub-cellular vesicles with a growing number of known biological functions. Microvesicles from a variety of parent cells within the vascular system increase in numerous pathological states. Red blood cell-derived MVs (RMVs) are relatively less studied than other types of circulating MVs despite red blood cells (RBCs) being the most abundant intravascular cell. This may be in part due the echoes of past misconceptions that RBCs were merely floating anucleate bags of hemoglobin rather than dynamic and responsive cells. The initial aim of this study was to maximize the concentration of RMVs derived from various blood or blood products by focusing on the optimal isolation conditions without creating more MVs from artificial manipulation. We found that allowing RBCs to sediment overnight resulted in a continuum in size of RBC membrane-containing fragments or vesicles extending beyond the 1 µm size limit suggested by many as the maximal size of an MV. Additionally, dilution and centrifugation factors were studied that altered the resultant MV population concentration. The heterogeneous size of RMVs was confirmed in mice models of hemolytic anemia. This methodological finding establishes a new paradigm in that it blurs the line between RBC, fragment, and RMV as well as suggests that the concentration of circulating RMVs may be widely underestimated given that centrifugation removes the majority of such RBC-derived membrane-containing particles. Full article
Show Figures

Figure 1

Figure 1
<p>Gentle sedimentation revealed vesicles larger than 1 µm in diameter. (<b>A</b>) Representative flow cytometry light scatter (forward scatter-area, or size, and side scatter-area, or internal complexity) dot plots of RBC-poor supernatant isolated by allowing newly-outdated (43–46 day old) donor blood to stand overnight at 4 °C, (<b>B</b>) with 3 µm beads added for size comparison. Staining with (<b>C</b>) anti-glycophorin-A antibodies and (<b>D</b>) lactadherin (which binds phosphatidylserine, PS) labeled (<b>E</b>) 97.5% ± 1.1% of events in 11 expired units’ RBC-poor supernatant. (<b>F</b>) Representative bright-field image of cells and vesicles in RBC-poor supernatant and (<b>G</b>) corresponding glycophorin-A fluorescent image showing relative similar fluorescence signal from the “macroparticles” of intact RBCs; arrows highlight vesicles larger than 1 µm (“macroparticles”). Bar = 10 µm.</p>
Full article ">Figure 2
<p>Size determination of cell-to-microparticle intermediate “macroparticles”: (<b>A</b>) Example identification of singlets in examining 2 µm beads using the forward scatter height by width; multiplets have disproportionately large width relative to height. (<b>B</b>) Light scatter of singlet beads (highlighted in the black circles) of various sizes was used to estimate the size of RBC macroparticles, with vertical lines corresponding to the forward scatter (FSC) of the singlet bead population. (<b>C</b>) Representative ungated RBC-poor supernatant with size windows shown by vertical lines corresponding to bead size based on data from (<b>B</b>). (<b>D</b>) This supernatant was examined for multiplets based on forward scatter height and width and gated to show (<b>E</b>) RBCs, (<b>F</b>) microparticles, (<b>G</b>) intermediately sized vesicles and possible microparticle doublets, and (<b>H</b>) macroparticles. Macroparticles made up 9.4% ± 9.3% of RBC-poor supernatant events smaller in diameter than 6 µm from units 1–4 days after expiration, and their diameter was 2.78 ± 0.35 µm based on their forward scatter height relative to that of the beads; <span class="html-italic">n</span> = 15 outdated units.</p>
Full article ">Figure 3
<p>Large vesicles were removed by centrifugation: To examine the effects of centrifugation on the macroparticle population, direct dilution was carried out. (<b>A</b>) The linear range of the cytometer was first established by minimizing the variance of a linear fit of the dilution to event rate; <span class="html-italic">n</span> = 3 donor units to identify the midpoint of the linear range. (<b>B</b>) Representative light scatter of PS+ or glycophorin A+ events comparing the resulting detected vesicle population. (i) With direct dilution, the RBC population overwhelmed the detection of vesicles (ii) seen with overnight sedimentation. (iii) Even centrifugation at 100× <span class="html-italic">g</span> for 10 min removed most large vesicles, and (iv) centrifugation at 1500× <span class="html-italic">g</span> for 10 min resulted in removal of over 85% of vesicles compared to overnight sedimentation. (<b>C</b>) There was a significant decrease in the forward scatter height of vesicles (&lt;6 µm) at 100× <span class="html-italic">g</span> or more, showing how the macroparticles are removed with low-speed centrifugation. (<b>D</b>) Compared to supernatant obtained from overnight sedimentation, there were 4.3-fold more vesicles in blood examined with the same settings used for the other supernatants. There was no significant difference in the total concentration of RBC vesicles at low speed (100 or 300× <span class="html-italic">g</span>), but there was a speed-dependent decrease in vesicles relative to supernatant from overnight sedimentation (inset); <span class="html-italic">n</span> = 8–11 (dilutions only had 8 units, whereas the different sedimentation/centrifugation numbers were performed on all 11). “1× <span class="html-italic">g</span>”/OS, overnight sedimentation; * <span class="html-italic">p</span> &lt; 0.05; ** denotes <span class="html-italic">t</span>-test <span class="html-italic">p</span>-val. &lt;0.001, and *** &lt;0.0001 relative to “1× <span class="html-italic">g</span>”/OS.</p>
Full article ">Figure 4
<p>Diluting blood before centrifugation enhances microparticles or microvesicles (MV/MP) sedimentation: The timing of dilution (either before or after centrifugation) was examined. (<b>A</b>) Aliquots of donor blood were centrifuged first, their supernatant removed and diluted 1:1, and then examined. <b>(B</b>) Identical aliquots of blood were first diluted 1:1, then centrifuged, and the supernatant examined. (<b>A</b>,<b>B</b>) Representative glycophorin A and FSC dot plots of two identical aliquots centrifuged for 5 min at 50× <span class="html-italic">g</span> then diluted or the reverse. Immediately prior to examination with flow cytometry, 3 µm counting beads were added to doubly confirm the cytometer reported volumes, and the dot plots displayed were of identical volumes (representative of 4 donor units). (<b>C</b>) Proof-of-principle showing as the blood became more diluted, centrifugation (for 1500× <span class="html-italic">g</span> for 15 min) resulted in significantly fewer RMVs; <span class="html-italic">n</span> = 3 (1 of the aforementioned 4 samples with the most MVs/MPs performed in triplicate).</p>
Full article ">Figure 5
<p>RBC MPs/RMVs sediment at lower speeds than PECAM-1 + MPs. As proof-of-principle, RBCs and platelet-rich plasma were subjected to extrusion through 0.45 µm pore size membranes to generate similarly sized MVs and, subsequently, centrifuged at different speeds before flow cytometry enumeration. (<b>A</b>) The relative size of the resulting MV populations (SSC-H) significantly decreased at all centrifugation speeds tested for RMVs, whereas there was no significant difference in the size of PECAM-1 + MVs until subjected to 1500× <span class="html-italic">g</span>. The green dotted line indicates the noise threshold of buffer-only liquid evaluated on the flow cytometer. (<b>B</b>) There was a significant drop in the number of RBC-MVs with any centrifugation speed; however, the decrease in PECAM-1 + MVs was not significant until 500× <span class="html-italic">g</span> or greater. (<b>C</b>) At 500× <span class="html-italic">g</span>, there were significantly fewer RMVs than PECAM-1 + MVs remaining relative to the initial unspun concentration. * Two-tailed <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05 compared to unspun; # <span class="html-italic">p</span> = 0.010 comparing the percent remaining at 500× <span class="html-italic">g</span>; results are technical triplicates from a single donor.</p>
Full article ">Figure 6
<p>Fluorescent lipid-labeled MPs/MVs adhered to RBCs. (<b>A</b>) To further investigate the discrepancy between RMV concentrations found in direct dilutions of blood compared to RBC-poor supernatant isolated by sedimenting cells overnight, isolated MVs were labeled with DiI and washed 3x (i). The 3rd wash buffer (with any unbound dye) was used to resuspend washed RBCs (ii). When the DiI-MVs were incubated with unlabeled washed cells, there appeared to be DiI-positive RBCs (iii). (<b>B</b>) Normalized to the total cells, there was a DiI-MV concentration-dependent increase in DiI-positive cells. (<b>C</b>) Light micrograph of an RBC, with corresponding (<b>D</b>) DiI-fluorescence and (<b>E</b>) overlay show apparent MV adhered to an otherwise normal-appearing RBC. Bar = 5 µm.</p>
Full article ">Figure 7
<p>Blood from mouse models of hemolytic anemia contained large and heterogeneously sized vesicles: (<b>A</b>) PECAM-1+ platelets and other MPs/MVs were excluded (red trapezoid exclusion gate) and (<b>B</b>) 3 µm beads were used to distinguish vesicles from cells (as murine RBCs are smaller than human). (<b>C</b>) Representative (PECAM-1 platelet/MV excluded) whole blood diluted 1:1000 in normal saline with 10% BSA and stained for TER119 in healthy control mice (C57BL/6), mice expressing solely human sickle hemoglobin (HbSS), and mice with deficient spectrin modeling hereditary spherocytosis. (<b>D</b>) Representative forward scatter height by width demonstrates heterogeneously sized RBC-MV intermediates in both sickle cell and spherocytosis mouse blood. (<b>E</b>) RBC vesicles smaller than 3 µm made up nearly 1% of all events in the sickle cell mouse blood and over 6% of all events in spherocytosis mouse blood; *** <span class="html-italic">p</span> &lt; 0.001 compared to normal mice; <span class="html-italic">n</span> = 3–5.</p>
Full article ">
Back to TopTop