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
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (3,326)

Search Parameters:
Keywords = exosomes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 1665 KiB  
Article
Exosomal mRNA Signatures as Predictive Biomarkers for Risk and Age of Onset in Alzheimer’s Disease
by Daniel A. Bolívar, María I. Mosquera-Heredia, Oscar M. Vidal, Ernesto Barceló, Ricardo Allegri, Luis C. Morales, Carlos Silvera-Redondo, Mauricio Arcos-Burgos, Pilar Garavito-Galofre and Jorge I. Vélez
Int. J. Mol. Sci. 2024, 25(22), 12293; https://doi.org/10.3390/ijms252212293 (registering DOI) - 15 Nov 2024
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline and memory loss. While the precise causes of AD remain unclear, emerging evidence suggests that messenger RNA (mRNA) dysregulation contributes to AD pathology and risk. This study examined exosomal mRNA expression [...] Read more.
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline and memory loss. While the precise causes of AD remain unclear, emerging evidence suggests that messenger RNA (mRNA) dysregulation contributes to AD pathology and risk. This study examined exosomal mRNA expression profiles of 15 individuals diagnosed with AD and 15 healthy controls from Barranquilla, Colombia. Utilizing advanced bioinformatics and machine learning (ML) techniques, we identified differentially expressed mRNAs and assessed their predictive power for AD diagnosis and AD age of onset (ADAOO). Our results showed that ENST00000331581 (CADM1) and ENST00000382258 (TNFRSF19) were significantly upregulated in AD patients. Key predictors for AD diagnosis included ENST00000311550 (GABRB3), ENST00000278765 (GGTLC1), ENST00000331581 (CADM1), ENST00000372572 (FOXJ3), and ENST00000636358 (ACY1), achieving > 90% accuracy in both training and testing datasets. For ADAOO, ENST00000340552 (LIMK2) expression correlated with a delay of ~12.6 years, while ENST00000304677 (RNASE6), ENST00000640218 (HNRNPU), ENST00000602017 (PPP5D1), ENST00000224950 (STN1), and ENST00000322088 (PPP2R1A) emerged as the most important predictors. ENST00000304677 (RNASE6) and ENST00000602017 (PPP5D1) showed promising predictive accuracy in unseen data. These findings suggest that mRNA expression profiles may serve as effective biomarkers for AD diagnosis and ADAOO, providing a cost-efficient and minimally invasive tool for early detection and monitoring. Further research is needed to validate these results in larger, diverse cohorts and explore the biological roles of the identified mRNAs in AD pathogenesis. Full article
(This article belongs to the Special Issue Molecular Advances in Alzheimer’s Disease 3.0)
Show Figures

Figure 1

Figure 1
<p>Volcano plots for mRNAs (<b>a</b>) conferring AD susceptibility, (<b>b</b>) differentially expressed mRNAs between the comparison groups, and (<b>c</b>) associated with ADAOO. Red lines show statistically significant mRNAs at 5%.</p>
Full article ">Figure 2
<p>Manhattan plots showing mRNA signatures (<b>a</b>) conferring susceptibility to AD (<span class="html-italic">p</span> &lt; 0.01 threshold, red line), (<b>b</b>) differentially expressed between study groups (<span class="html-italic">p</span> &lt; 2.5 × 10<sup>−6</sup> threshold, red line), and (<b>c</b>) associated with ADAOO (<span class="html-italic">p</span> &lt; 2.5 × 10<sup>−6</sup> threshold, red line) in a sample of 15 individuals with AD from Barranquilla, Colombia.</p>
Full article ">Figure 3
<p>(<b>a</b>) Accuracy and 95% confidence intervals for predicting AD diagnosis using different ML algorithms based on the top 30 mRNAs identified with OneR. (<b>b</b>) ROC curves for the xgbTree algorithm in the training (blue) and testing (green) datasets. (<b>c</b>) Variable importance analysis for the xgbTree algorithm. ROC: receiver operating characteristic; AUC: area under the ROC curve.</p>
Full article ">Figure 4
<p>Variable importance for the (<b>a</b>) rf, (<b>b</b>) xgbLinear, and (<b>c</b>) xgbTree ML algorithms for predicting ADAOO. Here, higher values are better.</p>
Full article ">
16 pages, 3484 KiB  
Article
Exosomes from Limosilactobacillus fermentum Ameliorate Benzalkonium Chloride-Induced Inflammation in Conjunctival Cells
by Kippeum Lee, Hyeonjun Gwon, Joo Yun Kim, Jae Jung Shim and Jae Hwan Lee
Int. J. Mol. Sci. 2024, 25(22), 12282; https://doi.org/10.3390/ijms252212282 - 15 Nov 2024
Abstract
Dry eye is characterized by persistent instability and decreased tear production, which are accompanied by epithelial lesions and inflammation on the surface of the eye. In our previous paper, we reported that supplementation with Limosilactobacillus fermentum HY7302 (HY7302) could inhibit corneal damage in [...] Read more.
Dry eye is characterized by persistent instability and decreased tear production, which are accompanied by epithelial lesions and inflammation on the surface of the eye. In our previous paper, we reported that supplementation with Limosilactobacillus fermentum HY7302 (HY7302) could inhibit corneal damage in a benzalkonium chloride (BAC)-induced mouse model of dry eye, through its effects in gut microbiome regulation. The aim of this study was to determine what functional extracellular substances can alter the inflammatory response of conjunctival cells. We isolated exosomes from HY7302 probiotic culture supernatant, analyzed their morphological characteristics, and found that their average size was 143.8 ± 1.1 nm, which was smaller than the exosomes from the L. fermentum KCTC 3112 strain. In addition, HY7302-derived exosomes significantly reduced the levels of genes encoding pro-inflammatory cytokines, including interleukin (IL)-20, IL-8, IL-6, and IL-1B, in BAC-treated human conjunctival cells. Moreover, HY7302-derived exosomes significantly increased the levels of genes encoding tight junction proteins, including TJP1, TJP2, and occludin-1, in Caco-2 cells. Lastly, the HY7302 exosomes reduced mRNA expression levels of IL1B, IL20, IL6, IL8, and NFAT5 in a transwell coculture system. Our findings indicate that HY7302 exosomes have potential for use in the treatment of ocular inflammation-related dry eye disease, through gut–eye axis communication via exosomes. Full article
(This article belongs to the Special Issue Molecular Advances in Dry Eye Syndrome)
Show Figures

Figure 1

Figure 1
<p>Images showing the morphology of exosomes from <span class="html-italic">Limosilactobacillus fermentum</span> HY7302 (HY7302). (<b>A</b>) HY7302 exosomes isolated by high-speed centrifugation. (<b>B</b>) Negative staining transmission electron microscopy images of exosomes isolated from <span class="html-italic">L. fermentum</span> HY7302. Scale bar, 50 nm. Arrowheads, exosomes.</p>
Full article ">Figure 2
<p>Characterization of exosomes from <span class="html-italic">Limosilactobacillus fermentum</span>. Nanoparticle tracking analysis of exosomes isolated from <span class="html-italic">L. fermentum</span> HY7302 (<b>A</b>) and KCTC3112 (<b>B</b>). Table box below show size distribution data obtained using a Malvern NanoSight NS300 and NanoSight NTA 3.4 Analytical software exosomes isolated from <span class="html-italic">L. fermentum</span> HY7302 (<b>C</b>) and KCTC3112 (<b>D</b>). Nanoparticle image of the isolated exosome from (<b>E</b>) HY7302 and (<b>F</b>) KCTC3112 were obtained using NTA analysis.</p>
Full article ">Figure 3
<p>Effect of exosomes from <span class="html-italic">Limosilactobacillus fermentum</span> HY7302 (7302E) on cytotoxicity. (<b>A</b>,<b>C</b>) Lactate dehydrogenase (LDH) release cytotoxicity assay and (<b>B</b>,<b>D</b>) 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test. Control cells or cells exposed to 0.0005% benzalkonium chloride (BAC) for 3 h were treated with <span class="html-italic">L. fermentum</span> HY7302 exosomes (7302E; 0, 0.01, 0.1, 0.5, 1.0, or 5.0 µg/10<sup>4</sup> cells). Data are expressed as mean ± standard deviation (SD) (n = 3). Different letters indicate significantly different values (<span class="html-italic">p</span> &lt; 0.05) (a &gt; ab &gt; b &gt; c).</p>
Full article ">Figure 4
<p>Effect of <span class="html-italic">Limosilactobacillus fermentum</span> HY7302 exosomes on tight junction molecules in Caco-2 cells. (<b>A</b>) Tight junction protein 1 (<span class="html-italic">TJP1)</span> and (<b>B</b>) occludin-1 were normalized to those of <span class="html-italic">GAPDH</span> and relative fold changes in their levels calculated. Levels of mRNA encoding (<b>C</b>) <span class="html-italic">TJP1</span>, (<b>D</b>) <span class="html-italic">TJP2</span>, and (<b>E</b>) occludin-1 were normalized to those of <span class="html-italic">GAPDH</span> and relative fold changes in their levels calculated. CON, control; HY7302, 10<sup>6</sup> CFU/mL HY7302; 7302EL, 0.5 μg/mL of HY7302 exosomes; 7302EH, 1 μg/mL of HY7302 exosomes. Data are expressed as mean ± standard deviation (SD) (n = 3). Different letters indicate significantly different values (<span class="html-italic">p</span> &lt; 0.05) (a &gt; ab &gt; b &gt; c &gt; d).</p>
Full article ">Figure 5
<p>Effects of <span class="html-italic">Limosilactobacillus fermentum</span> HY7302 extracellular vesicles (7302E) on pro-inflammatory factors in conjunctival cell lines treated using 0.0005% BAC. Levels of (<b>A</b>) interleukin-20 (<span class="html-italic">IL20</span>), (<b>B</b>) <span class="html-italic">IL8</span>, (<b>C</b>) <span class="html-italic">IL1B</span>, (<b>D</b>) <span class="html-italic">IL6</span>, (<b>E</b>) nuclear factor of activated T cells 5 (<span class="html-italic">NFAT5</span>), and (<b>F</b>) nuclear factor kappa B subunit 1 (<span class="html-italic">NFKB1</span>) mRNA were normalized to those of <span class="html-italic">GAPDH</span> and calculated as relative fold-change values. CON, control; BAC, 0.0005% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) BAC; 7302, 10<sup>6</sup> CFU/mL HY7302; 7302E, 1 μg/mL of HY7302 exosomes; 3112, 10<sup>6</sup> CFU/mL KCTC3112; 3112E, 1 μg/mL of KCTC3112 exosomes. Data are expressed as mean ± SD (n = 3). Different letters indicate significant differences (<span class="html-italic">p</span> &lt; 0.05) (a &gt; b &gt; c &gt; d).</p>
Full article ">Figure 6
<p>(<b>A</b>) Schematic of the experimental protocol to test the effect of exosomes of <span class="html-italic">Limosilactobacillus fermentum</span> HY7302 (7302E) on pro-inflammatory cytokines production. Clone 1-5c-4 cells were seeded in transwell plates and, once they reached confluence, exposed to Caco-2 cells. Caco-2 cells were co-cultured either with or without exosomes, using transwells, and culture medium samples of conjunctiva cell collected for analysis. Levels of (<b>B</b>) <span class="html-italic">IL1B</span>, (<b>C</b>) <span class="html-italic">IL20</span>, (<b>D</b>) <span class="html-italic">IL6</span>, (<b>E</b>) <span class="html-italic">IL8</span>, and (<b>F</b>) <span class="html-italic">NFAT5</span> mRNA in cells were normalized to those of <span class="html-italic">GAPDH</span> and relative fold-change values calculated. CON, control; BAC, 0.0005% (<span class="html-italic">v</span>/<span class="html-italic">v</span>) BAC; 7302, 10<sup>6</sup> CFU/mL HY7302; 7302E, 1 μg/mL of HY7302 exosomes; 3112, 10<sup>6</sup> CFU/mL KCTC3112; 3112E, 1 μg/mL of KCTC3112 exosomes. Data are expressed as mean ± SD (n = 3). Different letters indicate significant differences (<span class="html-italic">p</span> &lt; 0.05) (a &gt; ab &gt; b &gt; bc &gt; c).</p>
Full article ">
29 pages, 4005 KiB  
Article
Possible Role of Endothelial-Derived Cellular and Exosomal-miRNAs in Lipid-Mediated Diabetic Retinopathy: Microarray Studies
by Khaled Elmasry, Samar Habib, Inas Helwa, Mariam Lotfy Khaled, Ahmed S. Ibrahim, Amany Tawfik and Mohamed Al-Shabrawey
Cells 2024, 13(22), 1886; https://doi.org/10.3390/cells13221886 - 15 Nov 2024
Viewed by 32
Abstract
Diabetic retinopathy (DR) is a salient cause of blindness worldwide. There is still an immense need to understand the pathophysiology of DR to discover better diagnostic and therapeutic modalities. Human retinal endothelial cells (HRECs) were treated with 15-HETE or D-glucose, then miRNAs were [...] Read more.
Diabetic retinopathy (DR) is a salient cause of blindness worldwide. There is still an immense need to understand the pathophysiology of DR to discover better diagnostic and therapeutic modalities. Human retinal endothelial cells (HRECs) were treated with 15-HETE or D-glucose, then miRNAs were isolated, and a microarray was performed. MirWALK 2 and Ingenuity Pathway Analysis (IPA) were used to analyze the microarray results. Exosomal miRNAs from 15-HETE-treated HRECs were isolated, microarrayed, and then imported into IPA for further analysis. The microarray results showed that 15-HETE downregulated 343 miRNAs and upregulated 297 miRNAs in HRECs. High glucose treatment induced a differential expression of HREC-miRNAs where 185 miRNAs were downregulated and 244 were upregulated. Comparing the impact of 15-HETE versus DG or diabetic mouse retina elaborated commonly changing miRNAs. Pathway and target analysis for miRNAs changed in 15-HETE-treated HRECs revealed multiple targets and pathways that may be involved in 15-HETE-induced retinal endothelial dysfunction. The HREC-exosomal miRNAs were differentially expressed after 15-HETE treatment, with 34 miRNAs downregulated and 45 miRNAs upregulated, impacting different cellular pathways. Here, we show that 15-HETE induces various changes in the cellular and exosomal miRNA profile of HRECs, highlighting the importance of targeting the 12/15 lipoxygenase pathway in DR. Full article
(This article belongs to the Section Cells of the Cardiovascular System)
Show Figures

Figure 1

Figure 1
<p>Characterization of exosomes using Zetaview nanoparticle tracking (<b>A</b>), transmission electron microscopy (TEM) (<b>B</b>), and CD-63 immunogold labeling (<b>C</b>).</p>
Full article ">Figure 2
<p>Impact of 15-HETE on miRNAs profile of HRECs. Heat map demonstrating differential expression of miRNAs in control versus 15-HETE-treated HRECs. The blue color denotes downregulated miRNAs, and the red color signifies upregulated miRNAs (<b>A</b>). PCA mapping shows a distinction between different groups with less variation within the experiment (<b>B</b>). Pie chart demonstrating downregulated (<b>C</b>) and upregulated (<b>D</b>) miRNAs in HRECs in response to 15-HETE.</p>
Full article ">Figure 3
<p>High Glucose Treatment Induces alteration of the miRNA profile of HRECs. Heat map demonstrating differential expression of miRNAs in osmotic control versus high glucose-treated HRECs. The blue color denotes downregulated miRNAs, and the red color signifies upregulated miRNAs (<b>A</b>). PCA mapping shows a distinction between different groups with less variation within the experiment (<b>B</b>). Pie chart demonstrating downregulated (<b>C</b>) and upregulated (<b>D</b>) miRNAs in HRECs in response to high glucose treatment.</p>
Full article ">Figure 4
<p>Comparison of miRNAs commonly changed in HRECs challenged with 15-HETE for 24 h or HG for 5 days. Multiple common miRNAs that significantly changed under both conditions (<b>A</b>). miRNAs consistently altered in HRECs following treatment with 15-HETE or DG (<b>B</b>). IPA identified seven miRNAs that are commonly altered in both DG and 15-HETE-treated HRECs (<b>C</b>). IPA identified nine miRNAs commonly altered by 15-HETE-treated HRECs and diabetic mouse retinas (<b>D</b>).</p>
Full article ">Figure 5
<p>Mirwalk2 analysis of miRNAs in HRECs treated with 15-HETE for 24 h, demonstrating downregulated (<b>A</b>) and upregulated (<b>B</b>) miRNAs involved in DR as well as downregulated (<b>C</b>) and upregulated (<b>D</b>) miRNAs related to ER stress. Commonly downregulated (<b>E</b>) and upregulated (<b>F</b>) miRNAs are shown in Venn diagrams.</p>
Full article ">Figure 6
<p>miRNA microarray for RNA isolated from EVs released from HRECs treated with 15-HETE (0.1 µM) for 24 h. Heat map demonstrating differential expression of EV-derived miRNAs in control versus 15-HETE-treated HRECs. The blue color denotes downregulated miRNAs, and the red color signifies upregulated miRNAs (<b>A</b>). PCA mapping showing distinction between different groups with less variation within the experiment (<b>B</b>). Pie chart demonstrating significantly changed miRNAs (<b>C</b>). <span class="html-italic">p</span>-values are diagrammed (<b>D</b>).</p>
Full article ">
13 pages, 3002 KiB  
Article
Alterations in the Levels of Urinary Exosomal MicroRNA-183-5p and MicroRNA-125a-5p in Individuals with Type 2 Diabetes Mellitus
by Yixuan Fang, Shiyi Sun, Jing Wu, Guanjian Liu, Qinqin Wu and Xingwu Ran
Biomedicines 2024, 12(11), 2608; https://doi.org/10.3390/biomedicines12112608 - 14 Nov 2024
Viewed by 479
Abstract
Background: Type 2 diabetes mellitus (T2DM) is a metabolic disorder, and urinary exosomal microRNAs (miRNAs) were utilized as potential disease prediction or diagnostic biomarkers in numerous studies. This study investigated the differential expression of urinary exosomal miRNAs between non-diabetes mellitus (NDM) individuals and [...] Read more.
Background: Type 2 diabetes mellitus (T2DM) is a metabolic disorder, and urinary exosomal microRNAs (miRNAs) were utilized as potential disease prediction or diagnostic biomarkers in numerous studies. This study investigated the differential expression of urinary exosomal miRNAs between non-diabetes mellitus (NDM) individuals and those with T2DM. Aim: To elucidate the association between urinary exosomal miRNAs and T2DM. Methods: We recruited patients diagnosed with T2DM and NDM individuals in West China Hospital, Sichuan University, from November 2023 to February 2024. Subsequently, we performed sequencing of urinary exosomal microRNAs in both groups. The obtained sequencing results were further validated using RT-qPCR in both the training set and the validation set. Additionally, we conducted logistic regression analysis and Spearman correlation analysis on miRNAs with significant differential expression, as well as analysis of their biological functions. Results: A total of 118 urine samples were collected, 59 from individuals diagnosed with T2DM and 59 from NDM. There were differentially expressed miR-183-5p (p = 0.034) and miR-125a-5p (p = 0.008) between the two groups. Furthermore, multivariate regression analysis demonstrated that higher miR-125a-5p levels were negatively associated with the risk of T2DM (p = 0.044; OR: 0.046; 95% CI: 0.002, 0.922). Bioinformatics analysis indicated that the target genes of miR-183-5p were predominantly involved in insulin signaling and glucose transport processes, while those target genes of miR-125a-5p primarily mediated autophagy. Conclusions: miR-183-5p and miR-125a-5p might be involved in the pathogenesis of T2DM, while higher urinary exosomal miR-125a-5p was negatively associated with the risk of T2DM. Full article
(This article belongs to the Section Endocrinology and Metabolism Research)
Show Figures

Figure 1

Figure 1
<p>The process of screening participants.</p>
Full article ">Figure 2
<p>A comprehensive overview of the experimental design.</p>
Full article ">Figure 3
<p>miRNA profiling. (<b>A</b>) Volcano plot; green represents decreased miRNA levels, and red represents elevated miRNA levels in the T2DM group; (<b>B</b>) heat map.</p>
Full article ">Figure 4
<p>The RT-qPCR results of miRNAs; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01. (<b>A</b>) the result of miR-183-5p in training set; (<b>B</b>) the result of miR-29a-3p in training set; (<b>C</b>) the result of miR-125a-5p in training set; (<b>D</b>) the result of miR-183-5p in validation set; (<b>E</b>) the result of miR-125a-5p in validation set; NDM: non-diabetes mellitus; T2DM: type 2 diabetes mellitus.</p>
Full article ">Figure 5
<p>The scatter diagram and fitted curves of Spearman’s rank analysis. (<b>A</b>) The result of FPG and miR-125a-5p; (<b>B</b>) the result of HDLC and miR-125a-5p; (<b>C</b>) the result of HbA1c and miR-125a-5p. FPG, fasting plasma glucose; HDLC, high-density cholesterol; HbA1c, glycated hemoglobin A1c. The red line represents a fitted curve.</p>
Full article ">Figure 6
<p>Enrichment results of target genes corresponding to miR-183-5p and miR-125a-5p. (<b>A</b>) miR-183-5p KEGG pathway; (<b>B</b>) miR-125a-5p KEGG pathway.</p>
Full article ">Figure 7
<p>Enrichment results of target genes corresponding to miR-183-5p and miR-125a-5p. (<b>A</b>) miR-183-5p GO terms; (<b>B</b>) miR-125a-5p GO terms.</p>
Full article ">
20 pages, 1556 KiB  
Review
Spheroid-Exosome-Based Bioprinting Technology in Regenerative Medicine
by Hwa-Yong Lee and Jin Woo Lee
J. Funct. Biomater. 2024, 15(11), 345; https://doi.org/10.3390/jfb15110345 - 14 Nov 2024
Viewed by 207
Abstract
Since the discovery that exosomes can exchange genes, their potential use as tools for tissue regeneration, disease diagnosis, and therapeutic applications has drawn significant attention. Emerging three-dimensional (3D) printing technologies, such as bioprinting, which allows the printing of cells, proteins, DNA, and other [...] Read more.
Since the discovery that exosomes can exchange genes, their potential use as tools for tissue regeneration, disease diagnosis, and therapeutic applications has drawn significant attention. Emerging three-dimensional (3D) printing technologies, such as bioprinting, which allows the printing of cells, proteins, DNA, and other biological materials, have demonstrated the potential to create complex body tissues or personalized 3D models. The use of 3D spheroids in bioprinting facilitates volumetric tissue reconstruction and accelerates tissue regeneration via exosome secretion. In this review, we discussed a convergence approach between two promising technologies for bioprinting and exosomes in regenerative medicine. Among the various 3D cell culture methods used for exosome production, we focused on spheroids, which are suitable for mass production by bioprinting. We then summarized the research results on cases of bioprinting applications using the spheroids and exosomes produced. If a large number of spheroids can be supplied through bioprinting, the spheroid-exosome-based bioprinting technology will provide new possibilities for application in tissue regeneration, disease diagnosis, and treatment. Full article
(This article belongs to the Special Issue Advanced Technologies for Processing Functional Biomaterials)
Show Figures

Figure 1

Figure 1
<p>3D cell culture methods for exosome production.</p>
Full article ">Figure 2
<p>Bioprinting processes for exosome-related regenerative medicine: (<b>a</b>) Extrusion-based bioprinting, (<b>b</b>) Inkjet-based bioprinting, (<b>c</b>) Stereolithography bioprinting (Adapted with permission from Ref. [<a href="#B99-jfb-15-00345" class="html-bibr">99</a>]).</p>
Full article ">Figure 3
<p>(<b>a</b>) Three-dimensional bio-dot printing process with in situ formation of cell spheroids: (i) Schematic illustration showing the bio-dot printing procedure from PCL printing to induction of cell spheroids. (ii) Corresponding still images of the dot-printing process with PCL, matrix biomaterial ink, and cell-laden bio-ink (scale bars, 1 mm). (iii) Three-dimensional confocal images of constructs bio-dot-printed with red fluorescent micro-beads-laden sacrificial bio-ink at varying gelatin concentrations within matrix biomaterial ink: from 22.5 mg mL<sup>−1</sup> (G22.5) to 42.5 mg mL<sup>−1</sup> (G42.5). (iv) Microscopy images of HepG2 cells cultured for 3 days after bio-dot printing (scale bars, 200 μm). (v) Three-dimensional confocal images of cells stained with calcein-AM on day 0 and day 3 after printing (scale bars, 200 μm). (vi) SEM images of HepG2 spheroids produced through the conventional method and bio-dot printing process (scale bars, 200 μm). (vii) Histochemical and mmunocytochemical staining of the spheroids on day 5 (scale bars, 100 μm) (“Reprinted with permission from Ref. [<a href="#B83-jfb-15-00345" class="html-bibr">83</a>]. Copyright 2020 Wiley-VCH GmbH”). (<b>b</b>) Extrusion bioprinting process with the spheroid-loaded bioink. (i) Bioprinting method to form 3D constructs by the deposition of cell spheroids, suspended in a homogeneous printable hydrogel. (ii) The bioink was composed of mixtures of xanthan gum and alginate, capable of crosslinking with Ca<sup>2+</sup> ions. (iii) HMSC spheroids were obtained by culturing cells in a 3D microenvironment in non-cell-adhesive agarose hydrogel array inserts. (iv) By applying controlled pressure, the bioink was extruded through the nozzle in the form of a filament on pre-designed patterns to manufacture the constructs. (v) Constructs with proliferating cells were ionically crosslinked using calcium chloride and, subsequently, (vi) differentiated using a chondrogenic medium over 28 and 56 days to obtain chondral constructs. (vii) Typical visual aspect of 4-layered bioprinted constructs made of hMSC spheroids and XG3.75:A1.12 hydrogel. (viii) Spheroids maintained their 3D conformational structure 7 days after bioprinting. (ix) Size proportion between printed filaments and hMSC spheroid incorporated in the hydrogel 7 days after bioprinting. (x) Detachment of some cells from the 3D spheroid structure toward the hydrogel. (xi) In a clear trend, the open-pore structure was gradually reduced as the number of printed spheroids per construct increased (“Reprinted with permission from Ref. [<a href="#B126-jfb-15-00345" class="html-bibr">126</a>]. Copyright 2023 Wiley-VCH GmbH”).</p>
Full article ">
18 pages, 3068 KiB  
Article
Manganese Exposure Enhances the Release of Misfolded α-Synuclein via Exosomes by Impairing Endosomal Trafficking and Protein Degradation Mechanisms
by Dharmin Rokad, Dilshan S. Harischandra, Manikandan Samidurai, Yuan-Teng Chang, Jie Luo, Vivek Lawana, Souvarish Sarkar, Bharathi N. Palanisamy, Sireesha Manne, Dongsuk Kim, Gary Zenitsky, Huajun Jin, Vellareddy Anantharam, Auriel Willette, Arthi Kanthasamy and Anumantha G. Kanthasamy
Int. J. Mol. Sci. 2024, 25(22), 12207; https://doi.org/10.3390/ijms252212207 - 14 Nov 2024
Viewed by 266
Abstract
Excessive exposure to manganese (Mn) increases the risk of chronic neurological diseases, including Parkinson’s disease (PD) and other related Parkinsonisms. Aggregated α-synuclein (αSyn), a hallmark of PD, can spread to neighboring cells by exosomal release from neurons. We previously discovered that Mn enhances [...] Read more.
Excessive exposure to manganese (Mn) increases the risk of chronic neurological diseases, including Parkinson’s disease (PD) and other related Parkinsonisms. Aggregated α-synuclein (αSyn), a hallmark of PD, can spread to neighboring cells by exosomal release from neurons. We previously discovered that Mn enhances its spread, triggering neuroinflammatory and neurodegenerative processes. To better understand the Mn-induced release of exosomal αSyn, we examined the effect of Mn on endosomal trafficking and misfolded protein degradation. Exposing MN9D dopaminergic neuronal cells stably expressing human wild-type (WT) αSyn to 300 μM Mn for 24 h significantly suppressed protein and mRNA expression of Rab11a, thereby downregulating endosomal recycling, forcing late endosomes to mature into multivesicular bodies (MVBs). Ectopic expression of WT Rab11a significantly mitigated exosome release, whereas ectopic mutant Rab11a (S25N) increased it. Our in vitro and in vivo studies reveal that Mn exposure upregulated (1) mRNA and protein levels of endosomal Rab27a, which mediates the fusion of MVBs with the plasma membrane; and (2) expression of the autophagosomal markers Beclin-1 and p62, but downregulated the lysosomal marker LAMP2, thereby impairing autophagolysosome formation as confirmed by LysoTracker, cathepsin, and acridine orange assays. Our novel findings demonstrate that Mn promotes the exosomal release of misfolded αSyn by impairing endosomal trafficking and protein degradation. Full article
Show Figures

Figure 1

Figure 1
<p>Generation of GFP-tagged MN9D dopaminergic cells stably expressing human αSyn and characterization of exosomes. (<b>A</b>) Immunofluorescence of stably expressed GFP-fused human αSyn (red) in MN9D_αSynGFP cells, and GFP fluorescence (green) in both vector (control) MN9D_EVGFP cells and human αSyn-expressing MN9D_αSynGFP cells. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar, 100 µm. (<b>B</b>) Nanoparticle tracking analysis showing the concentration of exosomes from MN9D_αSynGFP cells from vehicle-stimulated (red) and Mn-stimulated (black) cells. (<b>C</b>) Nanoparticle tracking analysis showing the size distribution of exosomes from MN9D_αSynGFP cells from vehicle- (red) and Mn-stimulated (blue) cells. (<b>D</b>) TEM image of exosomes secreted from MN9D_αSynGFP cells displays distinctive morphology. Scale bar, 200 nm. (<b>E</b>) Western blots of GFP-fused human αSyn in exosomes from MN9D_αSynGFP cells compared to MN9D_EVGFP cells relative to exosome-positive markers Flotillin-1 and AIP-1/Alix in both cell types. (<b>F</b>) Average Thioflavin T (ThT) fluorescence in exosome samples isolated from Mn-treated and untreated αSyn-expressing and EV_MN9D cells, showing more aggregated αSyn in Mn-treated αSyn-expressing cells. (<b>G</b>) Protein aggregation rates (PAR) in exosome samples. Each trace and dot represent the average of 4 technical replicates. Statistically significant differences are denoted as * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 2–9.</p>
Full article ">Figure 2
<p>Mn exposure downregulates Rab11a expression both in vitro and in vivo. (<b>A</b>) Western blot of Rab11a from MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>B</b>) Densitometry analysis of Rab11a in Mn-exposed and control MN9D_αSynGFP cells. (<b>C</b>) ICC analysis of MN9D_αSynGFP cells (Rab11a; red) with and without Mn exposure for 24 h. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar, 50 µm. (<b>D</b>) Quantitative analysis of Rab11a integrated raw density from immunofluorescence analysis. (<b>E</b>) RT-qPCR analysis of <span class="html-italic">Rab11a</span> mRNA expression in MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>F</b>) Schematic illustration of Mn exposure in mice (30 mg/kg, male Swiss Webster and C57BL/6 mice) via oral gavage for 30 d. (<b>G</b>) Western blot of Rab11a from substantia nigral tissues from vehicle-treated and Mn-treated mice (30 mg/kg, male Swiss Webster). (<b>H</b>) Densitometry of Rab11a from substantia nigral tissues of vehicle- and Mn-exposed mice. (<b>I</b>) Western blot of control MN9D_αSynGFP (Vec) cells compared to the ectopic expression of wild-type (WT) Rab11a and dominant negative (DN) mutant Rab11a, respectively, in MN9D_αSynGFP cells transfected with WT Rab11a and DN Rab11 plasmids. (<b>J</b>) Concentration of exosomes from control and Mn-stimulated MN9D_αSynGFP cells (Vec), MN9D_αSynGFP cells expressing WT Rab11a, and MN9D_αSynGFP cells expressing DN Rab11a. (<b>K</b>) Nanoparticle tracking analysis showing the size distribution of exosome samples from exosome count, control and Mn-stimulated MN9D_αSynGFP cells (Vec; blue), MN9D_αSynGFP cells expressing WT Rab11a (green) and MN9D_αSynGFP cells expressing DN Rab11a (orange). Each group is represented by the mean ± S.E.M. from ≥3 separate measurements from vehicle- or Mn-treated groups. Statistically significant differences are denoted as * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001. <span class="html-italic">n</span> = 2–7.</p>
Full article ">Figure 3
<p>Mn exposure upregulates Rab27a expression both in vitro and in vivo. (<b>A</b>) Western blot of Rab27a in MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>B</b>) Densitometry of Rab27a in Mn-exposed and control MN9D_αSynGFP cells. (<b>C</b>) Immunofluorescence analysis of MN9D_αSynGFP cells (Rab27a; red) with and without Mn exposure for 24 h. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar, 50 µm. (<b>D</b>) Quantitative analysis of Rab27a integrated raw density from immunofluorescence analysis. (<b>E</b>) RT-qPCR analysis of <span class="html-italic">Rab27a</span> mRNA expression in MN9D_αSynGFP cells treated with and without Mn exposure for 24 h. (<b>F</b>) Western blot of Rab27a from substantia nigral tissues from vehicle- and Mn-treated mice (30 mg/kg for 30 days, male Swiss Webster). (<b>G</b>) Densitometry of Rab27a in substantia nigral tissues of vehicle- and Mn-exposed mice. Each group is represented by the mean ± S.E.M. from ≥6 separate measurements from vehicle- and Mn-treated groups. Statistically significant differences are denoted as * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. <span class="html-italic">n</span> = 4–10.</p>
Full article ">Figure 4
<p>Mn exposure impairs the autophagic/lysosomal system in MN9D_αSynGFP cells. (<b>A</b>) Western blot of Beclin 1, p62, and LAMP2 from MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>B</b>) Densitometry of Beclin 1 (<b>C</b>) p62 and (<b>D</b>) LAMP2 in vehicle- and Mn-exposed MN9D_αSynGFP cells. (<b>E</b>) Western blot of LC3B-I and II from MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>F</b>) Densitometry of LC3B-II in vehicle- and Mn-exposed MN9D_αSynGFP cells. (<b>G</b>) Immunofluorescence analysis of MN9D_αSynGFP cells (LAMP2; red) with and without Mn exposure for 24 h. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar, 50 µm. (<b>H</b>) Immunofluorescence analysis showing reduced LysoTracker intensity in MN9D_αSynGFP cells following Mn treatment. Scale bar, 50 µm. (<b>I</b>) Cathepsin assay showing increased activity following Mn treatment. (<b>J</b>) Acridine orange assay showing Mn exposure reduced signal intensities. (<b>K</b>) Western blot analysis of lysosomal marker LAMP2 and autophagic markers Beclin 1 and p62 in the substantia nigra of mice with and without Mn exposure (30 mg/kg for 30 days, male Swiss Webster). (<b>L</b>) Densitometry of Beclin 1, (<b>M</b>) p62, and (<b>N</b>) LAMP2 in the substantia nigra of mice with and without Mn exposure. Statistically significant differences are denoted as * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. <span class="html-italic">n</span> = 3–9.</p>
Full article ">Figure 5
<p>Mn exposure downregulates VPS35 expression in vitro. (<b>A</b>) Western blot of VPS35 from MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>B</b>) Densitometry analysis of VPS35 in Mn-exposed and control MN9D_αSynGFP cells. (<b>C</b>) ICC analysis of MN9D_αSynGFP cells (VPS35; red) with and without Mn exposure for 24 h. Nuclei were stained with Hoechst dye (blue). Magnification, 60×. Scale bar 50 µm. (<b>D</b>) Quantitative analysis of VPS35 integrated raw density from immunofluorescence analysis. (<b>E</b>) RT-qPCR analysis of VPS35 mRNA expression in MN9D_αSynGFP cells with and without Mn exposure for 24 h. (<b>F</b>) Western blot analysis of VPS35 in the substantia nigra of mice with and without Mn exposure (30 mg/kg for 30 days, male Swiss Webster). (<b>G</b>) Densitometry of VPS35 in the substantia nigra of mice with and without Mn exposure. Statistically significant differences are denoted as ** <span class="html-italic">p</span> &lt; 0.01. <span class="html-italic">n</span> = 2–8.</p>
Full article ">Figure 6
<p>Schematic representation of Mn’s potential to disrupt the endosomal trafficking and lysosomal pathway. Under normal, stress-free conditions, early endosomes form and are converted into late endosomes. Late endosomes can either be recycled back to the plasma membrane, as mediated by Rab11a, or they can mature into multivesicular bodies. MVBs either fuse with the plasma membrane, as mediated by Rab27a, to release exosomes, or MVBs can also merge with a lysosome to degrade its contents through the autophagolysosome pathway. Our study supports a model in which, when cells are exposed to Mn, key mediators of this endosomal trafficking pathway, such as Rab11a and Rab27a, as well as the lysosomal pathway, become dysregulated, leading to increased exosome release. Rab5 and Rab7 are involved in the maturation of early endosomes to late endosomes. Certain graphical representations in the above schematic depict proteins, such as divalent metal-ion transporter-1 (DMT-1), which are involved in Mn transport and homeostasis and are shown here in the shape of ion channels. The skull markings represent toxic conditions from Mn exposure.</p>
Full article ">
17 pages, 585 KiB  
Review
Select Endocrine Disorders and Exosomes in Early PDAC Diagnosis
by Barbara Wlodarczyk, Lukasz Durko, Konrad Walczak, Renata Talar-Wojnarowska and Ewa Malecka-Wojciesko
Int. J. Mol. Sci. 2024, 25(22), 12159; https://doi.org/10.3390/ijms252212159 - 13 Nov 2024
Viewed by 385
Abstract
Disturbances in carbohydrate metabolism are suggested to be the early symptoms of pancreatic ductal adenocarcinoma (PDAC). The accumulated data suggests that endocrine function-related biomarkers may represent a breakthrough in the early detection of PDAC. Factors which may predispose one to the development of [...] Read more.
Disturbances in carbohydrate metabolism are suggested to be the early symptoms of pancreatic ductal adenocarcinoma (PDAC). The accumulated data suggests that endocrine function-related biomarkers may represent a breakthrough in the early detection of PDAC. Factors which may predispose one to the development of PDAC are insulin resistance and hyperinsulinemia. Elevated insulin levels induce the onset of carcinogenesis by altering the differentiation and function of islet cells through stimulating growth factors, including insulin-like growth factors (IGFs). Impaired β cell function, along with the impact of PDAC-released factors (e.g., adrenomedullin (ADM), IGF-1, and macrophage inhibitory factor (MIF) on pancreatic islets, may contribute to the induction of diabetes associated with PDAC. Recently, exosomes have attracted worldwide attention due to their role in varied features of cell function, particularly in cancer progression. Exosomes comprise of small extracellular vesicles produced by almost all cells. These vesicles contain a vast array of biomolecules, including proteins and microRNAs. Exosomes participate in cancer growth and promote angiogenesis. They promote tumorigenesis and metastasis, and are associated with the acquisition of cancer cells resistant to chemotherapy. Data have been accumulating recently on the role of exosomes in the rapid recognition, prognosis and potential therapy of pancreatic cancer. Full article
(This article belongs to the Special Issue Pancreatic Cancer: Molecular Mechanisms and Therapeutic Strategies)
Show Figures

Figure 1

Figure 1
<p>The hypothesis on the role of NOD in PDAC development. Adrenomedullin (ADM), new-onset diabetes (NOD), pancreatic ductal adenocarcinoma (PDAC), endoplasmic reticulum kinase (ERK), and mitogen-activated protein kinases (MAPKs), ↓ means lower level/decrease.</p>
Full article ">
23 pages, 19767 KiB  
Article
Small and Long Non-Coding RNA Analysis for Human Trophoblast-Derived Extracellular Vesicles and Their Effect on the Transcriptome Profile of Human Neural Progenitor Cells
by Jessica A. Kinkade, Pallav Singh, Mohit Verma, Teka Khan, Toshihiko Ezashi, Nathan J. Bivens, R. Michael Roberts, Trupti Joshi and Cheryl S. Rosenfeld
Cells 2024, 13(22), 1867; https://doi.org/10.3390/cells13221867 - 11 Nov 2024
Viewed by 567
Abstract
In mice, the fetal brain is dependent upon the placenta for factors that guide its early development. This linkage between the two organs has given rise to the term, the placenta–brain axis. A similar interrelationship between the two organs may exist in humans. [...] Read more.
In mice, the fetal brain is dependent upon the placenta for factors that guide its early development. This linkage between the two organs has given rise to the term, the placenta–brain axis. A similar interrelationship between the two organs may exist in humans. We hypothesize that extracellular vesicles (EVs) released from placental trophoblast (TB) cells transport small RNA and other informational biomolecules from the placenta to the brain where their contents have pleiotropic effects. Here, EVs were isolated from the medium in which human trophoblasts (TBs) had been differentiated in vitro from induced pluripotent stem cells (iPSC) and from cultured iPSC themselves, and their small RNA content analyzed by bulk RNA-seq. EVs derived from human TB cells possess unique profiles of miRs, including hsa-miR-0149-3p, hsa-302a-5p, and many long non-coding RNAs (lncRNAs) relative to EVs isolated from parental iPSC. These miRs and their mRNA targets are enriched in neural tissue. Human neural progenitor cells (NPCs), generated from the same iPSC, were exposed to EVs from either TB or iPSC controls. Both sets of EVs were readily internalized. EVs from TB cells upregulate several transcripts in NPCs associated with forebrain formation and neurogenesis; those from control iPSC upregulated a transcriptional phenotype that resembled glial cells more closely than neurons. These results shed light on the possible workings of the placenta–brain axis. Understanding how the contents of small RNA within TB-derived EVs affect NPCs might yield new insights, possible biomarkers, and potential treatment strategies for neurobehavioral disorders that originate in utero, such as autism spectrum disorders (ASDs). Full article
(This article belongs to the Section Reproductive Cells and Development)
Show Figures

Figure 1

Figure 1
<p>Volcano plots for miRs and lncRNAs within EVs from TB vs. iPSC. Gray dots are miRs or lncRNAs that are not differentially expressed. (<b>A</b>) Volcano plot depicting the differential expression of microRNAs (miRs) within extracellular vesicles (EVs) from TB versus iPSC groups. Gray dots represent miRs that are not differentially expressed. Green dots indicate miRs with a log2 fold change difference between TB and iPSC groups, while red dots highlight miRs with both a significant log10 <span class="html-italic">p</span>-value and log2 fold change difference. (<b>B</b>) Volcano plot for long non-coding RNAs (lncRNAs) within EVs from TB versus iPSC groups. Gray dots denote non-differentially expressed lncRNAs. Green dots show lncRNAs with a log2 fold change between the two groups, and red dots represent lncRNAs with a significant log10 <span class="html-italic">p</span>-adjusted (<span class="html-italic">Q</span>)-value and log2 fold change difference.</p>
Full article ">Figure 2
<p>The analysis of individual miRs with the miRsTissueAtlas2 program [<a href="#B45-cells-13-01867" class="html-bibr">45</a>]. The diagram shows that hsa-miR0149-3p is predominantly expressed in the brain, hsa-miR-302a-5p is abundantly expressed in the heart, followed by the brain and nerve tissues, and hsa-miR-935 is almost exclusively expressed in the brain.</p>
Full article ">Figure 3
<p>TissueEnrich program [<a href="#B46-cells-13-01867" class="html-bibr">46</a>] analysis to determine which human organs and tissues have an abundance of transcripts that might be recognized by differentially expressed miRs shown in <a href="#cells-13-01867-f002" class="html-fig">Figure 2</a>. The primary mRNA targets for hsa-miR-0149-3p are enriched almost exclusively in the cerebral cortex. The mRNA targets for hsa-302a-5p are enriched in the cerebral cortex, followed by the prostate and thyroid gland. Primary mRNA targets for hsa-miR-395 are surprisingly abundant in the cervix and uterus.</p>
Full article ">Figure 4
<p>Extracellular vesicles (EVs) derived from human trophoblasts (TBs) and iPSCs and their internalization by human neural progenitor cells (NPCs). (<b>A</b>) A transmission electron microscopy (TEM) image of EVs derived from human TB cells. (<b>B</b>) A TEM image of EVs derived from human iPSCs.</p>
Full article ">Figure 5
<p>The internalization of EVs from TBs and iPSCs in human NPCs. (A) Fluorescence image of the internalization of EVs from human iPSCs. Red punctate material represents fluorescently tagged EVs (white arrows); the nuclei of NPCs are stained with DAPI (blue); and NPC fibers are labeled in green. (B) Fluorescence image of the internalization of EVs from human TB cells. Red punctate material represents fluorescently tagged EVs (white arrows); the nuclei of NPCs are stained with DAPI (blue); and NPC fibers are labeled in green.</p>
Full article ">Figure 6
<p>Transcriptome results of NPCs treated with TB EVs, iPSC EVs, and control NPCs. (<b>A</b>) A 2D PCA plot of NPCs treated with TB EVs (blue circles), iPSC EVs (green circles), and control NPCs (red circles). Clear separation is evident between control NPCs and those treated with TB EVs or iPSC EVs. (<b>B</b>) Heatmap analysis of NPCs treated with TB EVs, iPSC EVs, and control NPCs. The control NPC formed one cluster, whereas those treated with TB EVs and iPSC EVs showed some overlap between samples. (<b>C</b>) The volcano plot analysis of control NPCs vs. TB EVs treated with NPCs demonstrates several genes that show an increase of a more than 1.5-fold change (FC, shown in green), those few genes that have a −Log<sub>10</sub> <span class="html-italic">Q</span>-value (equivalent to <span class="html-italic">q</span> value ≤ 0.05, shown in light blue), and those that qualified both a −Log<sub>10</sub> <span class="html-italic">Q</span>-value and log<sub>2</sub> FC (shown in red). (<b>D</b>) The volcano plot analysis of control NPCs vs. iPSC EVs treated with NPCs demonstrates several genes that show an increase of more than 1.5-fold change (FC, shown in green), those few genes that have a −Log<sub>10</sub> <span class="html-italic">Q</span>-value (shown in light blue), and those that have qualified both a −Log<sub>10</sub> <span class="html-italic">Q</span>-value and log<sub>2</sub> FC (shown in red). Four independent replicates were assessed for each of the groups.</p>
Full article ">Figure 7
<p>STRING and hub gene analyses for proteins differentially expressed between control NPCs vs. TB EVs treated with NPCs. (<b>A</b>) Protein–protein interactions (PPI) were determined by STRING analysis. (<b>B</b>) The PPI files generated with STRING were imported into the cytoHubba app [<a href="#B67-cells-13-01867" class="html-bibr">67</a>] in Cytoscape [<a href="#B59-cells-13-01867" class="html-bibr">59</a>] to determine the top 10 hub proteins. Within this program, hub proteins were determined with MCC analysis as recommended [<a href="#B67-cells-13-01867" class="html-bibr">67</a>].</p>
Full article ">Figure 8
<p>STRING and hub gene analyses for proteins differentially expressed between control NPCs vs. iPSC EVs treated with NPCs. (<b>A</b>) Protein–protein interactions (PPI) were determined by STRING analysis. (<b>B</b>) The PPI.files generated with STRING were imported into the cytoHubba (Version 0.1) app [<a href="#B67-cells-13-01867" class="html-bibr">67</a>] in Cytoscape [<a href="#B59-cells-13-01867" class="html-bibr">59</a>] to determine the top 10 hub proteins. Within this program, hub proteins were determined with MCC analysis as recommended [<a href="#B67-cells-13-01867" class="html-bibr">67</a>].</p>
Full article ">Figure 9
<p>Gene ontology biological process (GO BP) and molecular function (GO MF) pathways are predicted to be affected based on differentially expressed genes. This was determined by using the WEB-based GEne SeT AnaLysis Toolkit (WebGestalt) 2019 version online program. (<b>A</b>) Control NPCs vs. TB EVs treated with NPCs. (<b>B</b>) Control NPCs vs. iPSC EVs.</p>
Full article ">Figure 10
<p>Brain-specific gene enrichment analysis for differentially expressed genes was determined by the GTEx Portal (API V2) [<a href="#B69-cells-13-01867" class="html-bibr">69</a>]. This was performed for the top 50 differentially expressed genes in each of the comparisons and by searching all brain regions in this database. (<b>A</b>) Control NPCs vs. TB EVs treated with NPCs. (<b>B</b>) Control NPCs vs. iPSCs treated with NPCs.</p>
Full article ">Figure 11
<p>Tissue enrichment analysis based on the TissueEnrich program [<a href="#B46-cells-13-01867" class="html-bibr">46</a>] with the 185 differentially expressed transcripts in human NPCs treated with TB-derived EVs that intersect with miR and lncRNA changes within EVs. (<b>A</b>) These transcripts are primarily associated with the placenta, followed by seminal vesicles, long, adipose tissue, the cerebral cortex, endometrium, ovary, gallbladder, cervix/uterine, and thyroid gland. (<b>B</b>) Heat map analysis reveals that the transcripts that are abundant in the placenta include <span class="html-italic">TMEM100</span>, <span class="html-italic">SVEP1</span>, <span class="html-italic">PTGES</span>, <span class="html-italic">PDGFB</span>, <span class="html-italic">PABPC4L</span>, <span class="html-italic">NRK</span>, <span class="html-italic">MSX2</span>, <span class="html-italic">MEOX2</span>, <span class="html-italic">HGF</span>, <span class="html-italic">DUSP9</span>, <span class="html-italic">CYTL1</span>, <span class="html-italic">CDKN1C</span>, and <span class="html-italic">APLN</span>.</p>
Full article ">
23 pages, 2299 KiB  
Review
Plant-Derived Exosome-like Nanoparticles: A Comprehensive Overview of Their Composition, Biogenesis, Isolation, and Biological Applications
by Ajia Sha, Yingyong Luo, Wenqi Xiao, Jing He, Xiaodie Chen, Zhuang Xiong, Lianxin Peng, Liang Zou, Bingliang Liu and Qiang Li
Int. J. Mol. Sci. 2024, 25(22), 12092; https://doi.org/10.3390/ijms252212092 - 11 Nov 2024
Viewed by 431
Abstract
Plant-derived exosome-like nanoparticles (PELNs) are a type of membranous vesicle isolated from plant tissues. They contain proteins, lipids, nucleic acids, and other components. PELNs are involved in the defensive response to pathogen attacks by exerting anti-inflammatory, antiviral, antifibrotic, and antitumor effects through the [...] Read more.
Plant-derived exosome-like nanoparticles (PELNs) are a type of membranous vesicle isolated from plant tissues. They contain proteins, lipids, nucleic acids, and other components. PELNs are involved in the defensive response to pathogen attacks by exerting anti-inflammatory, antiviral, antifibrotic, and antitumor effects through the substances they contain. Most PELNs are edible and can be used as carriers for delivering specific drugs without toxicity and side effects, making them a hot topic of research. Sources of PELNs are abundantly, and they can be produced in high yields, with a low risk of developing immunogenicity in vivo. This paper summarizes the formation, isolation, and purification methods; physical properties; and composition of PELNs through a comprehensive literature search. It also analyzes the biomedical applications of PELNs, as well as future research directions. This paper provides new ideas and methods for future research on PELNs. Full article
(This article belongs to the Special Issue The Molecular Basis of Extracellular Vesicles in Health and Diseases)
Show Figures

Figure 1

Figure 1
<p>Sources, biogenesis, and contents of PELNs. Route (1) shows the vacuole fusion with the PM to release the remaining ILVs, which are obtained from MVBs. Route (2) depicts the MVB fusion with the PM to release the ILVs as PELNs. Route (3) shows the EXPO secretion. Abbreviations: PM, cytoplasmic membrane; ER, endoplasmic reticulum; GA, Golgi apparatus; MVBs, multivesicular bodies; EXPO, extracellular-positive organelle. Partly based on literature mapping by Cui et al. [<a href="#B24-ijms-25-12092" class="html-bibr">24</a>] and Cong et al. [<a href="#B20-ijms-25-12092" class="html-bibr">20</a>].</p>
Full article ">Figure 2
<p>Isolation techniques for PELNs. (<b>a</b>) Ultracentrifugation; (<b>b</b>) Immunoprecipitation; (<b>c</b>) Polymer precipitation; (<b>d</b>) Size-exclusion chromatography; (<b>e</b>) Microfluidics.</p>
Full article ">Figure 3
<p>Biological applications of PELNs. Some of PELNs’ biological applications include anti-aging treatments [<a href="#B137-ijms-25-12092" class="html-bibr">137</a>], the treatment of periodontitis [<a href="#B138-ijms-25-12092" class="html-bibr">138</a>], drug delivery functions [<a href="#B137-ijms-25-12092" class="html-bibr">137</a>], the treatment of liver disease [<a href="#B139-ijms-25-12092" class="html-bibr">139</a>], the treatment of obesity [<a href="#B140-ijms-25-12092" class="html-bibr">140</a>], and the treatment of colon cancer [<a href="#B141-ijms-25-12092" class="html-bibr">141</a>].</p>
Full article ">
20 pages, 1822 KiB  
Article
Inert Gas Mild Pressure Action on Healthy Humans: The “IPA” Study
by Costantino Balestra, Clément Leveque, Simona Mrakic-Sposta, Mathias Coulon, Romain Tumbarello, Alessandra Vezzoli, Gerardo Bosco, Zuha Imtiyaz and Stephen R. Thom
Int. J. Mol. Sci. 2024, 25(22), 12067; https://doi.org/10.3390/ijms252212067 - 10 Nov 2024
Viewed by 237
Abstract
The goal of this study was to evaluate inflammatory and oxidative stress responses in human subjects (9 females and 15 males) (age [29.6 ± 11.5 years old (mean ± SD)], height [172.0 ± 10.05 cm], and weight [67.8 ± 12.4 kg]) exposed to [...] Read more.
The goal of this study was to evaluate inflammatory and oxidative stress responses in human subjects (9 females and 15 males) (age [29.6 ± 11.5 years old (mean ± SD)], height [172.0 ± 10.05 cm], and weight [67.8 ± 12.4 kg]) exposed to 1.45 ATA of helium (He) or nitrogen (N2) without concurrent hyperoxia. We hypothesized that elevated gas pressures would elicit an inflammatory response concurrent with oxidative stress. Consistent with ex vivo studies, both gasses elicited neutrophil activation, small elevations in microparticles (MPs) and increases in intra-MP interleukin (IL)-1β and inflammatory nitric oxide synthase, and an increase in urinary IL-6 concurrent with a marked reduction in plasma gelsolin. Mixed responses indictive of oxidative stress, with some biomarker elevations but little change in others and a decrease in some, were observed. Overall, these results demonstrate that exposure to typical diving gasses at a mildly elevated partial pressure will initiate inflammatory responses, which may play a significant role in decompression sickness (DCS). The complex pattern of oxidative stress responses may be indicative of competing systemic reactions and sampling different body fluids. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Activation of neutrophils after inert gas exposure (N<sub>2</sub> grey, He Blue). Data are shown as mean ± SD expressed in % of pre-exposure values of neutrophils (identified in flow cytometer based on CD66b expression) expressing myeloperoxidase (MPO) and CD18 above threshold value as index of cell activation (one-sample <span class="html-italic">t</span>-test and unpaired <span class="html-italic">t</span>-test). (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; ns = Non-Significant.)</p>
Full article ">Figure 2
<p>Microparticles in blood after helium or nitrogen exposure (N<sub>2</sub> grey, He Blue). Flow cytometry was used to evaluate MPs. Relative variations expressed in % of pre-exposure value of each that expressed proteins specific to different cells, including neutrophils (CD66b), endothelial cells (CD146), platelets (CD41a), and microglia (transmembrane protein 119, TMEM). As discussed in text, proteins expressing TSP-1 and F-actin, evaluated as those binding phalloidin, were also assessed. Data are shown as mean ± SD (* = <span class="html-italic">p</span> &lt; 0.05; ns = non-significant; <span class="html-italic">t</span>-test vs. control, with everyone acting as its own control (one-sample <span class="html-italic">t</span>-test)).</p>
Full article ">Figure 3
<p>IL-1B and iNOS (% of control values) (N<sub>2</sub> grey, He Blue). Data are shown as mean ± SD (** <span class="html-italic">p</span> &lt; 0.01; **** <span class="html-italic">p</span> &lt; 0.0001; ns = not significant; Wilcoxon and Mann–Whitney tests).</p>
Full article ">Figure 4
<p>Plasma gelsolin (% of control values) (N<sub>2</sub> grey, He Blue). Data are shown as mean ± SD (** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; Wilcoxon and Mann–Whitney tests; NS = not significant).</p>
Full article ">Figure 5
<p>Oxyinflammation % of control values (N<sub>2</sub> grey, He Blue)<b>.</b> Data are shown as mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; ns = not significant; <span class="html-italic">t</span>-test and one-sample <span class="html-italic">t</span>-test.</p>
Full article ">Figure 6
<p>Endothelial markers (N<sub>2</sub> grey, He Blue). VCAM-1. Data are shown as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; <span class="html-italic">t</span>-test and one-sample <span class="html-italic">t</span>-test.</p>
Full article ">Figure 7
<p>Experimental flowchart.</p>
Full article ">
16 pages, 4009 KiB  
Article
Berberine Mediates Exosomes Regulating the Lipid Metabolism Pathways to Promote Apoptosis of RA-FLS Cells
by Si-Fan Guo, Zhi-Bo Wang, Dan-Dan Xie, Ying Cai, Yan Wang, Xian Wang, Qiang Yang, Ai-Hua Zhang and Shi Qiu
Pharmaceuticals 2024, 17(11), 1509; https://doi.org/10.3390/ph17111509 - 9 Nov 2024
Viewed by 448
Abstract
Objectives: Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by joint damage and commonly linked to symptoms such as inflammation, swelling, and pain. Traditional Chinese Medicine offers complementary and integrative approaches in the management of rheumatoid arthritis, potentially providing additional options [...] Read more.
Objectives: Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by joint damage and commonly linked to symptoms such as inflammation, swelling, and pain. Traditional Chinese Medicine offers complementary and integrative approaches in the management of rheumatoid arthritis, potentially providing additional options that may help address treatment challenges and enhance overall patient care. This paper explores the mechanism of action of berberine from the perspective of cellular exosomes by mediating exosomal contents and thus treating RA. Methods: With the help of flow cytometry and confocal laser scanning microscope, it was determined that berberine promotes apoptosis in RA-FLS cells, and then lipid metabolomics technology was applied to screen and characterize the exosomes of RA-FLS cells to identify lipid core biomarkers closely related to RA, which were then projected into various databases for comprehensive analysis. Results: The data analysis showed that berberine could call back 11 lipid core biomarkers closely associated with RA, and interactive visualization of the database revealed that these markers were mainly focused on lipid metabolism aspects such as fatty acid elongation, degradation, and biosynthesis, as well as the biosynthesis of unsaturated fatty acids or PPARA activation of gene expression, PPARα‘s role in lipid metabolism regulation, glycerophospholipid metabolism, mitochondrial fatty acid oxidation disorders, and organelle biogenesis and maintenance. Conclusions: Berberine exerts its therapeutic effect on RA by mediating exosomal contents and thus regulating multiple lipid-related biological pathways, affecting the PPARγ-NF-κB complex binding rate, CREB and EGR-1 expression, cellular phagocytosis, and other aspects needed to inhibit proliferation and inflammatory responses in RA-FLS. This study offers a research foundation for exploring the mechanism of action of berberine in the treatment of RA. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structure of berberine.</p>
Full article ">Figure 2
<p>Different concentrations of berberine all promoted TNF-α-induced apoptosis of RA-FLS cells (model group, 25 μM, 50 μM, and 100 μM concentrations of berberine group) (<b>A</b>) Berberine cytotoxicity assay with CCK-8 reagent (<span class="html-italic">n</span> = 4 per group). (<b>B</b>) Analysis of Annexin V-FITC/PI staining in RA-FLS cells by flow cytometry (*** <span class="html-italic">p</span> &lt; 0.001) (n = 3 per group; all experiments were made in triplicate). (<b>C</b>) Representative images of RA-FLS apoptosis in the berberine group at different administered concentrations in the model group as detected by the Annexin V-FITC/PI assay (scale bar represents 20 μm, PI red, Annexin V-FITC green).</p>
Full article ">Figure 3
<p>Identification of differential metabolites in extracellular matrix of the model group and the berberine group. (<b>A</b>) Base peak ion chromatograms of mixed sample QC samples in positive ion mode and negative ion mode. (<b>B</b>) Volcanic map of differential metabolites. (Each dot in the volcanic diagram represents a metabolite, where purple dots represent down-regulated differential metabolites, red dots represent up-regulated differential metabolites, and gray represents detected metabolites with no significant difference). (<b>C</b>,<b>D</b>) The relative content charts of 11 lipid biomarkers in the model group and the berberine group as well as the violin chart of the relative content of a single substance were identified.</p>
Full article ">Figure 4
<p>Multivariate statistical analysis of differential metabolites. (<b>A</b>) PCA, (<b>B</b>) OPLS-DA, (<b>C</b>) scatter plot and OPLS-DA substitution test plot of 11 lipid biomarkers.</p>
Full article ">Figure 5
<p>Analysis of eleven lipid biomarkers combined with online databases. (<b>A</b>) Cluster analysis of eleven lipid biomarkers in the model group and the berberine group (M: model group; BER: berberine group). (<b>B</b>) Correlation analysis of eleven lipid biomarkers in the M group and the BER group (M: model group; BER: berberine group; pink is positively correlated; green is negatively correlated). (<b>C</b>) The depolarization sparse partial correlation algorithm (DSPC) module in the MetaboAnalyst network data processing platform analyzes the correlation of core biomarkers, and the more nodes, the higher the correlation. (<b>D</b>) Eleven lipid biomarkers were enriched and analyzed based on KEGG database. The color of the histogram represents the enrichment significance <span class="html-italic">p</span>-value, and the abscissa represents the enrichment ratio. (<b>E</b>) Eleven lipid biomarkers were enriched and analyzed based on the RaMP-DB database. The circle color represents the enrichment importance <span class="html-italic">p</span>-value, whereas the circle size shows the quantity of chemicals in the RaMP-DB pathway.</p>
Full article ">Figure 6
<p>The mechanism of berberine in the therapy of RA by mediating the contents of exosomes. (<b>A</b>) LPE lipids can competitively bind to PPARγ with NF-κB. Berberine inhibits the separation of the PPARγ-NF-κB complex by up-regulating the content of LPE in exosomes, forming a protein–protein interaction and controlling the expression of the NF-κB pathway, thus affecting the proliferation of FLS. (<b>B</b>) Palmitic acid can promote the release of chemokines and cytokines that cause inflammation and stimulate the activation of CREB and EGR-1; the over-expression of EGR-1 activates the genes encoding α1 and α2 chains of type I collagen, which leads to synovial fibrosis. Berberine can affect the above process by down-regulating the content of palmitic acid. (<b>C</b>) The compound 1-(9Z-octadecenoyl)-sn-glycerol-3-phosphocholine can be transformed into PtdSer, which activates GTPase Rac and phagocytizes dying cells. Berberine can accelerate the phagocytosis of dying cells by up-regulating the content of 1-(9Z-octadecenoyl)-sn-glycerol-3-phosphocholine, thus affecting the progress of RA. (Note: (<b>A</b>) Light yellow and yellow represent cytoplasm and nucleus; blue stands for protein; green represents the signal path; purple stands for LPE substance. (<b>B</b>) Light yellow and yellow represent the cell membrane and nucleus; green, blue, and orange represent proteins; Pink represents TNF-α; Colored circles represent proinflammatory factors; Green and purple concentric circles represent palmitic acid. (<b>C</b>) Orange stands for enzyme).</p>
Full article ">
16 pages, 3435 KiB  
Article
Harnessing a Safe Novel Lipid Nanoparticle: Targeted Oral Delivery to Colonic Epithelial and Macrophage Cells in a Colitis Mouse Model
by Rabeya Jafrin Mow, Michal Pawel Kuczma, Xiaodi Shi, Sridhar Mani, Didier Merlin and Chunhua Yang
Nanomaterials 2024, 14(22), 1800; https://doi.org/10.3390/nano14221800 - 9 Nov 2024
Viewed by 400
Abstract
A novel lipid nanoparticle (nLNP), formulated with three essential lipids to mimic ginger-derived exosomal particles, shows strong potential for delivering IL-22 mRNA specifically to the colon, presenting a unique oral drug delivery system for inflammatory bowel disease (IBD). However, its cellular targets and [...] Read more.
A novel lipid nanoparticle (nLNP), formulated with three essential lipids to mimic ginger-derived exosomal particles, shows strong potential for delivering IL-22 mRNA specifically to the colon, presenting a unique oral drug delivery system for inflammatory bowel disease (IBD). However, its cellular targets and uptake behavior in healthy versus diseased colons remain unclear. Understanding these aspects is crucial for fully elucidating its targeting effectiveness in inflamed colon tissue. This study investigates the nLNP’s cellular targets in healthy and diseased mouse colons. Flow cytometry compared nLNP uptake in healthy mice and a DSS-induced acute colitis model. The results revealed efficient internalization of nLNP by colonic epithelial cells in healthy and inflamed mice. In non-inflamed mice, the small number of colonic macrophages resulted in minimal uptake of nLNP by these cells. In inflamed mice, macrophages migrated to the damaged epithelium, where nLNP uptake was significantly increased, highlighting the nLNP’s ability to target both epithelial and macrophage cells during inflammation. Additionally, safety assessments showed that the nLNP neither altered in vitro kinase activities nor exhibited immunotoxicity or induced in vivo toxicity at the maximum tolerated oral dose. These findings underscore the nLNP’s safety and potential as a promising epithelial/macrophage-targeted drug delivery platform for oral ulcerative colitis (UC) treatment. Full article
Show Figures

Figure 1

Figure 1
<p>Characterization of nLNP. (<b>A</b>–<b>C</b>) Representative images showing size, zeta potential, and atomic force microscopy (AFM) results for pristine nLNP. (<b>D</b>–<b>F</b>) Representative images showing size, zeta potential, and AFM results for DiA-labeled nLNP.</p>
Full article ">Figure 2
<p>Biodistribution of DiR-tagged nLNP in mouse organs after oral administration. The representative picture shows the fluorescence detected in different mouse organs (stomach, heart, brain, small intestine, lung, kidney, spleen, liver, caecum, and colon).</p>
Full article ">Figure 3
<p>Acute colitis decreases epithelial cell numbers while increasing lamina propria cells. (<b>A</b>) Pie charts showing the distribution of cells isolated from the epithelium compared to the lamina propria layer of colon tissues. (<b>B</b>) Bar graph comparing the ratio of epithelial cells (EC) to lamina propria cells (LP) with and without inflammation. Error bars represent one standard deviation (n = 3; * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Proportions of immune cells (CD45+), epithelial cells (CD45-EpCAM+), and macrophages (CD45+CD11b+F4/80+) in cells isolated from the epithelium and lamina propria. (<b>A</b>) FACS analysis of immune cells from healthy (left) and inflamed (right) epithelium. (<b>B</b>) Comparison of immune cell proportions in the epithelium between healthy and diseased colon. (<b>C</b>) FACS analysis of macrophages from healthy (left) and inflamed (right) epithelium. (<b>D</b>) Comparison of macrophage proportions in the epithelium between healthy and diseased colon. (<b>E</b>) FACS analysis of immune cells from healthy (left) and inflamed (right) lamina propria. (<b>F</b>) Comparison of immune cell proportions in the lamina propria between healthy and diseased colon. (<b>G</b>) FACS analysis of macrophages from healthy (left) and inflamed (right) lamina propria. (<b>H</b>) Comparison of macrophage proportions in the lamina propria between healthy and diseased colon. Error bars represent one standard deviation (n = 3; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 5
<p>Increased nLNP uptake by macrophages in the epithelial fraction of inflamed mice. (<b>A</b>) t-SNE analysis of FACS data shows the proportion of epithelial cells (EpCAM+DiA+) and macrophages (CD45+CD11b+DiA+) that uptake DiA-labeled nLNP in healthy (<b>upper</b>) and inflamed (<b>lower</b>) tissue. (<b>B</b>) Comparison of macrophages internalizing nLNP between healthy and diseased colon epithelium. Error bars represent one standard deviation (n = 3; * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>FACS analysis of cultured PBMCs, gating on viable CD4+ and CD8+ T cells in solvent control (SC) and nLNP-treated groups, with (+) or without (−) IL-2 addition. (<b>A</b>) A representative FACS image shows the percentage of viable proliferating CD4+ and CD8+ T cells in the SC group without IL-2 addition. (<b>B</b>) Percentage of viable proliferating CD4+ and CD8+ T cells in the nLNP-treated group without IL-2 addition. (<b>C</b>) Comparison of proliferated viable T cells between the nLNP-treated and SC groups without IL-2 addition. (<b>D</b>) Percentage of viable proliferating CD4+ and CD8+ T cells in SC group with IL-2 addition. (<b>E</b>) Percentage of viable proliferating CD4+ and CD8+ T cells in the nLNP-treated group with IL-2 addition. (<b>F</b>) Comparison of proliferated viable T cells between the nLNP-treated and SC group with IL-2 addition. Data are presented as mean ± SD (n = 3, ns: non-significant).</p>
Full article ">Figure 7
<p>Hematological and biochemical analysis of CD-1 mice from Control and nLNP groups (n = 4). Blood was collected from the retro-orbital sinus, and 25–50 μL of whole blood was used for hematological analyses (VetScan HM5; Abaxis, CA, USA), and 100 µL was used for biochemical analysis (VetScan VS2; Abaxis, CA, USA). The following hematologic parameters are shown: (<b>A</b>) Blood counts and hemoglobin: WBC-white blood cells; RBC-red blood cells; HGB-hemoglobin; (<b>B</b>) Liver enzymes: ALT-alanine aminotransferase; ALP-alkaline phosphatase; TBIL-total bilirubin; (<b>C</b>) Biochemical parameters: BUN-urea nitrogen; ALB-albumin. All results are shown as means ± SD (n = 4, ns: non-significant).</p>
Full article ">
21 pages, 1576 KiB  
Review
Corneal Treatment, Repair, and Regeneration: Exosomes at Rescue
by Brooke T. Robbins, Kate A. Montreuil, Neloy Kundu, Prashant Kumar and Vibhuti Agrahari
Pharmaceutics 2024, 16(11), 1424; https://doi.org/10.3390/pharmaceutics16111424 - 7 Nov 2024
Viewed by 444
Abstract
Exosomes are extracellular vesicles within the nanosized range that play roles in intercellular communication and thus have certain biological activities. The secretory signaling communication mechanism is an efficient way of exchanging information between cells and has been investigated as nature’s therapeutic drug carriers. [...] Read more.
Exosomes are extracellular vesicles within the nanosized range that play roles in intercellular communication and thus have certain biological activities. The secretory signaling communication mechanism is an efficient way of exchanging information between cells and has been investigated as nature’s therapeutic drug carriers. This review will summarize the potential of exosomes as therapeutic tools and drug delivery vehicles for corneal pathologies. The cornea is an avascular ocular tissue, and its healing is a complex process including cell death and migration, cell proliferation and differentiation, and extracellular matrix remodeling. Here, we discussed the structure, barrier, phases, and healing cascade of cornea. We briefly reviewed the immunogenicity and toxicity of exosomes and role of exosomes in preserving cornea. Additionally, we provided combining exosome strategies with hydrogels, gene and stem cells therapy focused on corneal treatment, repair, and regeneration. Full article
Show Figures

Figure 1

Figure 1
<p>Layers of cornea (prepared using Google drawings).</p>
Full article ">Figure 2
<p>Four phases of epithelial wound-healing (prepared using Google drawings).</p>
Full article ">Figure 3
<p>Cascade during corneal injury and inflammatory responses (prepared using Google drawings).</p>
Full article ">Figure 4
<p>Biogenesis of MSC-derived exosomes. (<b>a</b>) comprises of endocytosis, MVB formation, and exosomes secretion into the extracellular environment through merging with the plasma membrane. (<b>b</b>) represents contents and characteristics of exosomes carrying a variety of substances such as proteins, lipids, nucleic acids, metabolites [<a href="#B26-pharmaceutics-16-01424" class="html-bibr">26</a>].</p>
Full article ">
22 pages, 4075 KiB  
Article
Supplementation with a Whey Protein Concentrate Enriched in Bovine Milk Exosomes Improves Longitudinal Growth and Supports Bone Health During Catch-Up Growth in Rats
by Jorge García-Martínez, Rafael Salto, María D. Girón, Íñigo M. Pérez-Castillo, Pilar Bueno Vargas, Jose D. Vílchez, Azahara Linares-Pérez, Manuel Manzano, María T. García-Córcoles, Ricardo Rueda and José M. López-Pedrosa
Nutrients 2024, 16(22), 3814; https://doi.org/10.3390/nu16223814 - 7 Nov 2024
Viewed by 422
Abstract
Background: Undernutrition impairs linear growth while restoration of nutritional provisions leads to accelerated growth patterns. However, the composition of the nutrition provided is key to facilitating effective catch-up growth without compromising bone quantity, quality, and long-term health. Methods: We evaluated the role of [...] Read more.
Background: Undernutrition impairs linear growth while restoration of nutritional provisions leads to accelerated growth patterns. However, the composition of the nutrition provided is key to facilitating effective catch-up growth without compromising bone quantity, quality, and long-term health. Methods: We evaluated the role of a whey protein concentrate enriched in bovine milk exosomes (BMEs) in modulating the proliferative properties of human chondrocytes in vitro and studied how these effects might impact bone quantity and quality measured as longitudinal tibia growth, bone mineral content (BMC) and density (BMD), and trabecular micro-CT parameters in stunted rats during catch-up growth. Results: BMEs promoted proliferation in C28/I2 human chondrocytes mediated by mTOR-Akt signaling. In a stunting rat model, two-week supplementation with BMEs during refeeding was associated with improved tibia BMD, trabecular microstructure (trabecular number (Tb. N.) and space (Tb. Sp.)), and a more active growth plate (higher volume, surface, and thickness) compared to non-supplemented stunted rats. Positive effects on physis translated to significantly longer tibias without compromising bone quality when extending the refeeding period for another two weeks. Conclusions: Overall, BME supplementation positively contributed to longitudinal bone growth and improved bone quantity and quality during catch-up growth. These findings might be relevant for improving diets aimed at addressing the nutritional needs of children undergoing undernutrition during early life. Full article
(This article belongs to the Special Issue Nutritional Supplements for Bone Health)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Research model of diet-induced catch-up growth. BME—group refed with the diet supplemented with whey protein concentrate enriched in bovine milk exosomes; CTR—group refed with the control diet; NR—non-restricted group; and RR—restricted group.</p>
Full article ">Figure 2
<p>The effects of BMEs on the growth of human chondrocytes. (<b>a</b>) Bar plot showing the effects of bovine milk exosomes (0–50 µg/mL; 72 h incubation) on C28/I2 metabolic activity in the MTT test (<span class="html-italic">n</span> = 6); (<b>b</b>) impact of sonication on bovine milk exosome biological function in a repeated MTT test (0–50 µg/mL; 48 h incubation) (<span class="html-italic">n</span> = 4). Data represented as mean ± SD. BME—whey protein concentrate enriched in bovine milk exosomes. * <span class="html-italic">p</span>-value &lt; 0.05 compared to the control (0 µg/mL); <sup>#</sup> <span class="html-italic">p</span>-value &lt; 0.05 compared to non-sonicated bovine milk exosomes.</p>
Full article ">Figure 3
<p>The effects of BMEs on cell cycle profile in human chondrocytes. Control: 0 and 24 h incubation; BME: 15 µg/mL, 24 h incubation. Data represented as mean (<span class="html-italic">n</span> = 10). BME—whey protein concentrate enriched in bovine milk exosomes. * <span class="html-italic">p</span>-value &lt; 0.05 compared to the control.</p>
Full article ">Figure 4
<p>The effects of BMEs on clone formation in human chondrocytes. Data represented as mean ± SD (<span class="html-italic">n</span> = 10). BME—whey protein concentrate enriched in bovine milk exosomes. * <span class="html-italic">p</span>-value &lt; 0.05 compared to the control.</p>
Full article ">Figure 5
<p>The effects of BMEs on intracellular signaling mechanisms. Bar plots showing the effects of BME and sonicated BME on (<b>a</b>) Akt, (<b>b</b>) ERK1/2, (<b>c</b>) AMPK, (<b>d</b>) mTOR, and (<b>e</b>) p70S6K; (<b>f</b>) representative western blot images of signaling components. Control: 0 µg/mL, 24 h; BME/Sonicated BME: 30 µg/mL, 24 h. Data represented as mean ± SD. BME—whey protein concentrate enriched in bovine milk exosomes. * <span class="html-italic">p</span>-value &lt; 0.05 compared to the control; <sup>#</sup> <span class="html-italic">p</span>-value &lt; 0.05 compared to non-sonicated bovine milk exosomes.</p>
Full article ">Figure 6
<p>The effects of BME incubation (30 µg/mL, 24 h) on the expression of genes involved in bone formation and development. (<b>a</b>) Gene Ontology (GO) functional enrichment analysis results; (<b>b</b>) genes at least 2-fold-overexpressed following BME incubation. Color codes gene ontology evidence can be consulted in reference [<a href="#B43-nutrients-16-03814" class="html-bibr">43</a>]. Data represented as mean ± SD. BME—whey protein concentrate enriched in bovine milk exosomes.</p>
Full article ">Figure 7
<p>Representative images (<b>a</b>) and micro-CT parameters (<b>b</b>–<b>f</b>) of tibia trabecular structure upon completion of the two-week refeeding period. Data represented as mean ± SD. BME—group refed with the diet supplemented with whey protein concentrate enriched in bovine milk exosomes; BV/TV—bone volume to total volume ratio; Conn. Dens.—connective density; CTR—group refed with the control diet; RR—restricted group; Tb. N.—trabecular number; Tb. Sp.—trabecular separation; and Tb. Th.—trabecular thickness. * <span class="html-italic">p</span>-value &lt; 0.05 compared to the control refeeding group (CTR); <sup>#</sup> <span class="html-italic">p</span>-value &lt; 0.05 compared to the restricted group (RR).</p>
Full article ">Figure 8
<p>Representative scans (<b>a</b>) and micro-CT parameters (<b>b</b>–<b>d</b>) of the tibia growth plate upon completion of the two-week refeeding period. Data represented as mean ± SD. BME—group refed with the diet supplemented with whey protein concentrate enriched in bovine milk exosomes; CTR—group refed with the control diet; GP—growth plate; and RR—restricted group. * <span class="html-italic">p</span>-value &lt; 0.05 compared to the control refeeding group (CTR); <sup>#</sup> <span class="html-italic">p</span>-value &lt; 0.05 compared to the restricted group (RR).</p>
Full article ">Figure 9
<p>Growth velocity of the experimental and control animals. Data represented as mean ± SD. BME—group refed with the diet supplemented with whey protein concentrate enriched in bovine milk exosomes; CTR—group refed with the control diet. * <span class="html-italic">p</span>-value &lt; 0.05 compared to the control refeeding group (CTR).</p>
Full article ">Figure 10
<p>The effects of the four-week refeeding period on total body (<b>b</b>) and tibia length (<b>a</b>). Data represented as mean ± SD. BME—group refed with the diet supplemented with whey protein concentrate enriched in bovine milk exosomes; CTR—group refed with the control diet. * <span class="html-italic">p</span>-value &lt; 0.05 compared to the control refeeding group (CTR).</p>
Full article ">
14 pages, 1143 KiB  
Review
The Coordinated Changes in Platelet Glycan Patterns with Blood Serotonin and Exosomes
by Fusun Kilic
Int. J. Mol. Sci. 2024, 25(22), 11940; https://doi.org/10.3390/ijms252211940 - 6 Nov 2024
Viewed by 249
Abstract
The structures of glycans, specifically their terminal positions, play an important role as ligands for receptors in regulating the adhesion ability of platelets. Recent advances in our understanding of free/unbound serotonin (5-HT) in blood plasma at supraphysiological levels implicate it as one of [...] Read more.
The structures of glycans, specifically their terminal positions, play an important role as ligands for receptors in regulating the adhesion ability of platelets. Recent advances in our understanding of free/unbound serotonin (5-HT) in blood plasma at supraphysiological levels implicate it as one of the most profound influencers in remodeling the platelet’s surface N-glycans. Proteomic analysis of the membrane vesicles identified enzymes, specifically glycosyltransferases, only on the surface of the platelets isolated from the supraphysiological level of 5-HT-containing blood plasma. However, these enzymes can only be effective on the cell surface under certain biological conditions, such as the level of their substrates, temperature, and pH of the environment. We hypothesize that exosomes released from various cells coordinate the required criteria for the enzymatic reaction on the platelet surface. The elevated plasma 5-HT level also accelerates the release of exosomes from various cells, as reported. This review summarizes the findings from a wide range of literature and proposes mechanisms to coordinate the exosomes and plasma 5-HT in remodeling the structures of N-glycans to make platelets more prone to aggregation. Full article
(This article belongs to the Special Issue Emerging Roles for Serotonin in Regulating Metabolism)
Show Figures

Figure 1

Figure 1
<p>The density of SERT molecules on the plasma membrane exhibits a biphasic relationship to plasma 5-HT concentration ([5-HT]). An initial increase in [5-HT upregulates the density of SERT on platelet surface. However, a further increase in plasma [5-HT] downregulates the density of SERT molecules [<a href="#B13-ijms-25-11940" class="html-bibr">13</a>,<a href="#B46-ijms-25-11940" class="html-bibr">46</a>]. Indeed, our in vivo [<a href="#B13-ijms-25-11940" class="html-bibr">13</a>] and in vitro [<a href="#B46-ijms-25-11940" class="html-bibr">46</a>] studies confirm a dynamic relationship between extracellular 5-HT elevation, loss of surface SERT, and depletion of platelet 5-HT [<a href="#B45-ijms-25-11940" class="html-bibr">45</a>,<a href="#B48-ijms-25-11940" class="html-bibr">48</a>].</p>
Full article ">Figure 2
<p>Proposed model for crosstalk between 5-HT receptors and SERT. In platelets, 5-HT signaling is mediated through 5-HT2A and G protein-coupled Gq receptors. These receptors transduce the signals via independent, but interconnected, pathways to promote platelet aggregation. The activation of 5-HT2A receptor signaling activates phospholipase C (PLC) and results in the hydrolysis of phosphatidylinositol 4,5-biphosphate (PIP2) to inositol-1,4,5-triphosphate (IP3) [<a href="#B17-ijms-25-11940" class="html-bibr">17</a>,<a href="#B25-ijms-25-11940" class="html-bibr">25</a>,<a href="#B27-ijms-25-11940" class="html-bibr">27</a>,<a href="#B28-ijms-25-11940" class="html-bibr">28</a>]. IP3 activates the serine/threonine protein kinase C and facilitates Ca<sup>2+</sup> mobilization. Elevated free Ca<sup>2+</sup> in the cytoplasm activates transglutaminase (TGase), which modifies Rab4/Rho/Rac with 5-HT (serotonylation) [<a href="#B9-ijms-25-11940" class="html-bibr">9</a>,<a href="#B25-ijms-25-11940" class="html-bibr">25</a>,<a href="#B27-ijms-25-11940" class="html-bibr">27</a>,<a href="#B28-ijms-25-11940" class="html-bibr">28</a>,<a href="#B29-ijms-25-11940" class="html-bibr">29</a>,<a href="#B30-ijms-25-11940" class="html-bibr">30</a>,<a href="#B31-ijms-25-11940" class="html-bibr">31</a>,<a href="#B44-ijms-25-11940" class="html-bibr">44</a>,<a href="#B45-ijms-25-11940" class="html-bibr">45</a>,<a href="#B46-ijms-25-11940" class="html-bibr">46</a>,<a href="#B48-ijms-25-11940" class="html-bibr">48</a>]. The Rab4 family of small GTPases regulates vesicular traffic [<a href="#B30-ijms-25-11940" class="html-bibr">30</a>,<a href="#B31-ijms-25-11940" class="html-bibr">31</a>,<a href="#B48-ijms-25-11940" class="html-bibr">48</a>]. I propose that in platelets, Rab4 is associated with early endosomes and regulates membrane recycling. In its active form (GTP bound), Rab4 facilitates the exocytosis of small vesicles that use the cytoskeletal network for their translocation to the plasma membrane. The translocation of small vesicles brings the glycosylation enzymes to the platelet surface.</p>
Full article ">
Back to TopTop