[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (482)

Search Parameters:
Keywords = sialic acids

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 1255 KiB  
Article
Capillary Electrophoresis-Laser Induced Fluorescence Method Development and Validation for Quantification of Nine Gangliosides—Application to Analysis of Cell Lines of CNS Origin
by Katinka Tarnóczi, Orsolya Geda, Tamás Tábi and Éva Szökő
Molecules 2024, 29(16), 3769; https://doi.org/10.3390/molecules29163769 - 9 Aug 2024
Viewed by 274
Abstract
Gangliosides are sialic acid-containing glycosphingolipids that play an essential role in many biological and pathophysiological processes. They are present in high amounts in the central nervous system and their abnormal metabolism or expression has been observed in many diseases. We have developed and [...] Read more.
Gangliosides are sialic acid-containing glycosphingolipids that play an essential role in many biological and pathophysiological processes. They are present in high amounts in the central nervous system and their abnormal metabolism or expression has been observed in many diseases. We have developed and validated a sensitive capillary electrophoresis laser-induced fluorescence (CE-LIF) method for the separation and quantification of oligosaccharides digested from nine gangliosides of high biological relevance. APTS was used for the labeling of the glycans. Reverse polarity CE was performed for the separation of the labeled glycans bearing negative charges. The optimized background electrolyte is a 15 mM lithium acetate buffer with pH of 5 containing 5% w/v linear polyacrylamide, which allows for the separation of all nine gangliosides. Validation parameters including linearity, precision, and accuracy were evaluated. LOQ and LOD were in the nM range, comparable to those of LC-MS techniques. The method was used to identify and quantify the ganglioside pattern of glioblastoma and neuroblastoma cell lines. The presented method is a valuable tool for further investigations aiming at understanding the role of gangliosides in various neurological diseases or CNS tumors. Full article
Show Figures

Figure 1

Figure 1
<p>Structure of the nine gangliosides most abundant in biological samples [<a href="#B4-molecules-29-03769" class="html-bibr">4</a>].</p>
Full article ">Figure 2
<p>Effect of various buffer pH (<b>A</b>) and LiAc buffer concentration (<b>B</b>) on the separation of the glycans of nine gangliosides. Separation voltage: −25 kV; temperature: 25 °C; capillary: 50/60 cm × 75 µm i.d. uncoated fused silica capillary; injection 0.5 psi/5 s.</p>
Full article ">Figure 3
<p>Glycan separation of ganglioside spiked glioblastoma cell extracts under optimized separation conditions. The cells were spiked with 1250 nM ganglioside standards each, except GQ1b, which was 625 nM. Separation buffer: 15 mM LiAc, pH 5.0, containing 5% <span class="html-italic">w</span>/<span class="html-italic">v</span> LPA. Separation voltage: −25 kV; temperature: 25 °C; capillary: 50/60 cm × 75 µm i.d. uncoated fused silica capillary; injection 0.5 psi/5 s.</p>
Full article ">Figure 4
<p>Electropherogram of ganglioside extract glycans from C6 glioblastoma cell line sample. Separation buffer: 15 mM LiAc, pH 5.0, containing 5% <span class="html-italic">w</span>/<span class="html-italic">v</span> LPA. Separation voltage: −25 kV; temperature: 25 °C; capillary: 50/60 cm × 75 µm i.d. uncoated fused silica capillary; injection 0.5 psi/5 s.</p>
Full article ">Figure 5
<p>Electropherogram of ganglioside extract glycans from SH-SY5Y neuroblastoma cell line sample. Separation buffer: 15 mM LiAc, pH 5.0, containing 5% <span class="html-italic">w</span>/<span class="html-italic">v</span> LPA. Separation voltage: −25 kV; temperature: 25 °C; capillary: 50/60 cm × 75 µm i.d. uncoated fused silica capillary; injection 0.5 psi/5 s.</p>
Full article ">
20 pages, 3939 KiB  
Article
Structural Characterization and Abundance of Sialylated Milk Oligosaccharides in Holstein Cows during Early Lactation
by Lisa Isernhagen, Christina E. Galuska, Andreas Vernunft and Sebastian P. Galuska
Foods 2024, 13(16), 2484; https://doi.org/10.3390/foods13162484 - 7 Aug 2024
Viewed by 307
Abstract
Among other bioactive molecules, milk contains high amounts of sialylated milk oligosaccharides (MOs) that influence numerous processes in the offspring. For instance, sialylated MOs inhibit the invasion of pathogens and positively influence the gut microbiome to support the optimal development of the offspring. [...] Read more.
Among other bioactive molecules, milk contains high amounts of sialylated milk oligosaccharides (MOs) that influence numerous processes in the offspring. For instance, sialylated MOs inhibit the invasion of pathogens and positively influence the gut microbiome to support the optimal development of the offspring. For these reasons, sialylated MOs are also used in infant formula as well as food supplements and are potential therapeutic substances for humans and animals. Because of the high interest in sialylated bovine MOs (bMOs), we used several analytical approaches, such as gas and liquid chromatography in combination with mass spectrometry, to investigate in detail the profile of sialylated bMOs in the milk of Holstein Friesian cows during early lactation. Most of the 40 MOs identified in this study were sialylated, and a rapid decrease in all detected sialylated bMOs took place during the first day of lactation. Remarkably, we observed a high variance within the sialylation level during the first two days after calving. Therefore, our results suggest that the content of sialylated MOs might be an additional quality marker for the bioactivity of colostrum and transitional milk to ensure its optimized application for the production of milk replacer and food supplements. Full article
Show Figures

Figure 1

Figure 1
<p>Quantification of Neu5Gc in milk from Holstein Friesian cows. (<b>A</b>) Sampling timeline for milk collection. The time ranges of colostrum, transitional milk, and mature milk were specified on the basis of Silva et al. [<a href="#B49-foods-13-02484" class="html-bibr">49</a>]. Created with BioRender.com. (<b>B</b>) Scheme of the DMB-RP-HPLC strategy for sialic acid quantification. Sialic acid residues in milk as well as in the extracted bMOs fraction were released by hydrolysis and subsequently labeled with DMB for fluorescence detection using an RP-HPLC system equipped with a fluorescence detector. Created with BioRender.com. (<b>C</b>) Box and whisker plots (median; min to max) showing the Neu5Gc values during early lactation (n = 6 animals) as well as the values for Neu5Ac (<b>D</b>) and Neu5Gc (<b>E</b>) in the extracted bMOs fraction (n = 5 animals). The statistical analysis and graphs were generated using BioRender.com. Significant differences are denoted as follows: *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, ***: <span class="html-italic">p</span> &lt; 0.001, ****: <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Analysis of bMO distribution using LC-MS. (<b>A</b>) Scheme of bMO analysis using LC-MS. After bMO enrichment using PGC cartridges, the resulting bMOs were analyzed using HILIC-HESI-MS/(MS). Created with BioRender.com. (<b>B</b>) The displayed MS fragments of bMOs were used for the identification of bMOs. For the manual verification process, at least one of those fragments had to be found in the corresponding MS<sup>2</sup> spectrum to identify the detected bMO. For the sialylated bMOs, the most significant fragment was always the cleaved sialic acid residues (Neu5Ac or Neu5Gc). The bMO structures were computed with GlycoWorkbench 2 [<a href="#B43-foods-13-02484" class="html-bibr">43</a>].</p>
Full article ">Figure 3
<p>Proposed structures of the detected disaccharides lactose and lactosamine in addition to bMOs in Holstein Friesian cows during early lactation. For some bMOs, multiple linkages and compositions have been reported. Due to the lack of specific linkage analysis and for clarity purposes, not all possible isomers are visualized. The bMO structures were designed with GlycoWorkbench 2 [<a href="#B43-foods-13-02484" class="html-bibr">43</a>] and assembled using BioRender.com. Each structure is given the corresponding number used in this study, which is also described in <a href="#foods-13-02484-t001" class="html-table">Table 1</a>.</p>
Full article ">Figure 4
<p>LC-MS analysis of sialyllactose (SL). (<b>A</b>) Extracted ion chromatogram (EIC) of SLs shown for representative samples on the day of calving. (<b>B</b>) MS spectrum at 5.46 min showing the deprotonated molecular ion [M-H]<sup>−</sup> of SL with its corresponding MS/MS spectrum (<b>D</b>) showing two MO fragments as a cross-ring fragment of a Hex with 87.0072 <span class="html-italic">m</span>/<span class="html-italic">z</span> and a detached Neu5Ac at 290.0884 <span class="html-italic">m</span>/<span class="html-italic">z</span>. (<b>C</b>) MS spectrum at 6.68 min showing the deprotonated molecular ion [M-H]<sup>−</sup> of SL with its corresponding MS/MS spectrum (<b>E</b>) showing fragments at 87.0072 and 290.0883 <span class="html-italic">m</span>/<span class="html-italic">z</span>, as well as Neu5Ac attached to a Hex (470.1522 <span class="html-italic">m</span>/<span class="html-italic">z</span>) and Neu5Ac attached to a Hex with an additional cross-ring fragment of another Hex (572.1846 <span class="html-italic">m</span>/<span class="html-italic">z</span>). The bMO structures were designed with GlycoWorkbench 2 [<a href="#B43-foods-13-02484" class="html-bibr">43</a>].</p>
Full article ">Figure 5
<p>LC-MS analysis of DSL. (<b>A</b>) MS spectrum at 10.99 min showing the deprotonated molecular ion for DSL containing a Neu5Ac dimer attached to the lactose core and the corresponding MS<sup>2</sup> spectrum for DSL at this time. The MS<sup>2</sup> spectrum shows three fragments, two of which are already mentioned for SL in <a href="#foods-13-02484-f004" class="html-fig">Figure 4</a> (87.0072 and 290.0881 <span class="html-italic">m</span>/<span class="html-italic">z</span>), and one larger fragment is SL with 632.2120 <span class="html-italic">m</span>/<span class="html-italic">z</span>, validating the linear composition of DSL. (<b>B</b>) MS spectrum at 11.51 min showing the deprotonated molecular ion for heterogenic DSL (hDSL) consisting of Neu5Ac as well as Neu5Gc attached to the lactose core and the corresponding MS<sup>2</sup> spectrum for hDSL at this time. The MS<sup>2</sup> spectrum shows four fragments, two of which were already mentioned for SL in <a href="#foods-13-02484-f004" class="html-fig">Figure 4</a> (87.0071 and 290.0885 <span class="html-italic">m</span>/<span class="html-italic">z</span>), as well as one larger fragment, NGL, with 648.2013 <span class="html-italic">m</span>/<span class="html-italic">z</span>, validating the linear composition of hDSL as NGL with an additional Neu5Ac attached. (<b>C</b>) The MS spectrum at 12.69 min displays the deprotonated molecular ion for hDSL and the corresponding MS<sup>2</sup> spectrum for hDSL at this time. The MS<sup>2</sup> spectrum shows four fragments, three of which were already mentioned for hDSL (87.0071, 290.0872, and 306.0837 <span class="html-italic">m</span>/<span class="html-italic">z</span>), as well as SL with 632.2031 <span class="html-italic">m</span>/<span class="html-italic">z</span>, validating the linear composition of the second hDSL as SL with an additional Neu5Gc attached. The bMO structures were designed with GlycoWorkbench 2 [<a href="#B43-foods-13-02484" class="html-bibr">43</a>].</p>
Full article ">Figure 6
<p>LC-MS analysis of sialyllactosamine (SLN). (<b>A</b>) Extracted ion chromatogram (EIC) of SLN shown for a representative sample on the day of calving. (<b>B</b>) MS spectrum at 5.10 min showing the deprotonated molecular ion 673.2306 <span class="html-italic">m</span>/<span class="html-italic">z</span> of SLN with the corresponding MS<sup>2</sup> spectrum, (<b>D</b>) which displays the same fragments as those displayed for SL in <a href="#foods-13-02484-f005" class="html-fig">Figure 5</a>, with an additional fragment of another cross-ring fragment of Hex at 170.0449 <span class="html-italic">m</span>/<span class="html-italic">z</span>. (<b>C</b>) MS spectrum at 5.50 min showing the deprotonated molecular ion 673.2311 <span class="html-italic">m</span>/<span class="html-italic">z</span> of SLN with the corresponding MS<sup>2</sup> spectrum, (<b>E</b>) which shows the same fragments as noted at 5.10 min. Additionally, there is another fragment of Neu5Ac attached to a cross-ring fragment of a Hex (306.1190 <span class="html-italic">m</span>/<span class="html-italic">z</span>), which indicates the α2,6-linkage of Neu5Ac. The bMO structures were computed with GlycoWorkbench 2 [<a href="#B43-foods-13-02484" class="html-bibr">43</a>].</p>
Full article ">Figure 7
<p>Distribution of bMOs during lactation. (<b>A</b>) The total ion chromatography (TIC) from the colostrum samples on day 0 p.p. of the analyzed Holstein cattle were overlaid using FreeStyle software (Thermo Fisher). The MO peaks were labeled with the name and retention time of the base peak. (<b>B</b>) The TIC from the colostrum samples on day 30 p.p. of the analyzed Holstein cattle were overlaid using FreeStyle software. The MO peaks were labeled with the name and retention time of the base peak. The ratios of the peak areas of the sialylated bMOs were determined (n = 5 animals), and box and whisker plots (median; min to max) are shown for (<b>C</b>) day 0 and (<b>D</b>) day 30 p.p. Only sialylated bMOs with peak areas greater than 0.1% were included. The statistical analysis and graphs were generated using BioRender.com. Significant differences are denoted as follows: *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, ***: <span class="html-italic">p</span> &lt; 0.001, ****: <span class="html-italic">p</span> &lt; 0.0001. The bMO structures were designed with GlycoWorkbench 2 [<a href="#B43-foods-13-02484" class="html-bibr">43</a>].</p>
Full article ">Figure 8
<p>Abundance of the major sialylated bMOs during early lactation. The EICs were used to calculate the ratios of the peak areas of the most abundant sialylated bMOs. Box and whisker plots (median; min to max) are shown for (<b>A</b>) 3′-SL, (<b>B</b>) DSL, (<b>C</b>) 3′SLN, and (<b>D</b>) 6′-SL (n = 5 animals). Statistical analysis and graphs were generated using BioRender.com. Significant differences are denoted as follows: *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, ***: <span class="html-italic">p</span> &lt; 0.001, ****: <span class="html-italic">p</span> &lt; 0.0001. The bMO structures were designed with GlycoWorkbench 2 [<a href="#B43-foods-13-02484" class="html-bibr">43</a>].</p>
Full article ">
16 pages, 3813 KiB  
Article
Structural Characterization of Human Bufavirus 1: Receptor Binding and Endosomal pH-Induced Changes
by Mitchell Gulkis, Mengxiao Luo, Paul Chipman, Mario Mietzsch, Maria Söderlund-Venermo, Antonette Bennett and Robert McKenna
Viruses 2024, 16(8), 1258; https://doi.org/10.3390/v16081258 - 6 Aug 2024
Viewed by 392
Abstract
Bufaviruses (BuV) are members of the Parvoviridae of the Protoparvovirus genus. They are non-enveloped, T = 1 icosahedral ssDNA viruses isolated from patients exhibiting acute diarrhea. The lack of treatment options and a limited understanding of their disease mechanisms require studying these viruses [...] Read more.
Bufaviruses (BuV) are members of the Parvoviridae of the Protoparvovirus genus. They are non-enveloped, T = 1 icosahedral ssDNA viruses isolated from patients exhibiting acute diarrhea. The lack of treatment options and a limited understanding of their disease mechanisms require studying these viruses on a molecular and structural level. In the present study, we utilize glycan arrays and cell binding assays to demonstrate that BuV1 capsid binds terminal sialic acid (SIA) glycans. Furthermore, using cryo-electron microscopy (cryo-EM), SIA is shown to bind on the 2/5-fold wall of the capsid surface. Interestingly, the capsid residues stabilizing SIA binding are conserved in all human BuVs identified to date. Additionally, biophysical assays illustrate BuV1 capsid stabilization during endo–lysosomal (pH 7.4–pH 4) trafficking and capsid destabilization at pH 3 and less, which correspond to the pH of the stomach. Hence, we determined the cryo-EM structures of BuV1 capsids at pH 7.4, 4.0, and 2.6 to 2.8 Å, 3.2 Å, and 2.7 Å, respectively. These structures reveal capsid structural rearrangements during endo–lysosomal escape and provide a potential mechanism for this process. The structural insights gained from this study will add to the general knowledge of human pathogenic parvoviruses. Furthermore, the identification of the conserved SIA receptor binding site among BuVs provides a possible targetable surface-accessible pocket for the design of small molecules to be developed as anti-virals for these viruses. Full article
Show Figures

Figure 1

Figure 1
<p>Glycan array of BuV1 demonstrates sialic acid binding. (<b>A</b>) Relative fluorescence units for each of the 600 glycans are shown with the standard deviation in red. The numbers of the 15 glycans with the highest signal are displayed, with the top 5 glycans shown in larger text. (<b>B</b>) Table showing the symbols of the glycan structure of the top five glycans with the highest signal. Pink diamonds represent N-acetylneuraminic acid, yellow circles represent galactose, blue squares represent N-acetylglucosamine, and green circles represent mannose. Glycosidic linkages are indicated between sugars.</p>
Full article ">Figure 2
<p>BuV1 utilizes sialic acid for cell adhesion. (<b>A</b>) Glycan symbols show the glycans expressed on the surface of Pro5 cells (pink), Lec2 cells (yellow), and Lec8 cells (blue). Pink diamonds represent N-acetylneuraminic acid, yellow circles represent galactose, blue squares represent N-acetylglucosamine, and green circles represent mannose. The rightmost glycan is the terminal glycan. (<b>B</b>) Bar graphs show the percentage of fluorescently labeled cells compared to the total cell count. Bars are colored as in (<b>A</b>).</p>
Full article ">Figure 3
<p>Structural characterization of BuV1 in complex with sialic acid. (<b>A</b>) SDS-PAGE gel of purified BuV1 capsids at pH 7.4. (<b>B</b>) Representative micrograph of BuV1 VLPs embedded in vitreous ice. A scale bar is given in the lower left corner. (<b>C</b>) 2.16 Å reconstructed electron density map shown with a sharpening B factor of −90 at 2σ. The surface is colored radially, as indicated in the scale bar to the right. Locations of bound sialic acid are shown in purple. (<b>D</b>) Cartoon structure of a BuV1 monomer is shown in pink. The bound sialic acid is shown in stick form in purple. The monomer is shown with regions participating in icosahedral symmetry elements on the exterior labeled with blue shapes. The 5-fold axis is denoted by the pentagon, the 3-fold by the triangle, and the 2-fold by the ellipse. (<b>E</b>) The interacting pocket is shown in detail with residues (pink) labeled, which hydrogen bond with the sialic acid (purple). Electron density is shown for the sialic acid at 1σ with a sharpening B factor of −50 Å<sup>2</sup>.</p>
Full article ">Figure 4
<p>BuV1 shows greater thermal stability at lower pHs. (<b>A</b>) Representative traces of the first derivative of measured fluorescence are shown for differential scanning fluorimetry experiments conducted at pH 7.4 (green), pH 6.0 (orange), pH 5.5 (red), and pH 4.0 (teal). (<b>B</b>) Graph, which shows melting temperature as a function of pH. (<b>C</b>) Table showing the melting temperature (T<sub>m</sub>) of BuV1 at different pHs. Each experiment was performed on three separate days, with three technical repeats on each day.</p>
Full article ">Figure 5
<p>Reconstruction of BuV1 capsids at pH 7.4, 4.0, and 2.6. (<b>A</b>–<b>C</b>, <b>left</b>) Representative micrographs are shown for BuV1 at pH 7.4 (<b>A</b>), 4.0 (<b>B</b>), and 2.6 (<b>C</b>). A scale bar is given in the lower left corner. A partial capsid is indicated with a red arrow (<b>C</b>). (<b>A</b>–<b>C</b>, <b>middle</b>) Sharpened reconstructed electron density maps for BuV1 at pH 7.4 (<b>A</b>), 4.0 (<b>B</b>), and 2.6 (<b>C</b>) are shown at 2σ. The surface is colored radially, as indicated in the scale bar to the far right. (<b>A</b>–<b>C</b>, <b>right</b>) Zoomed view of the 5-fold and 2-fold symmetry axes for the reconstructed electron density maps for BuV1 at pH 7.4 (<b>A</b>), 4.0 (<b>B</b>), and 2.6 (<b>C</b>) shown at 2σ. The surface is colored radially, as indicated in the scale bar to the far right.</p>
Full article ">Figure 6
<p>BuV1 monomers at pH 7.4, 4.0, and 2.6 reveals pH-dependent differences. (<b>A</b>) Overlay of a monomer from the final 60-mer models of the BuV1 capsid at pH 7.4 (green), 4.0 (teal), and 2.6 (tan). The monomers are aligned using secondary structure matching to the BuV1 pH 7.4 monomer. The monomer is shown with regions participating in icosahedral symmetry elements on the exterior labeled with blue shapes. The 5-fold axis is denoted by the pentagon, the 3-fold by the triangle, and the 2-fold by the ellipse. (<b>B</b>) Zoomed view of the differences in the N-terminal observed between models of the BuV1 capsid at pH 7.4 (green), 4.0 (teal), and 2.6 (tan). Ordering of 10 additional residues is observed at pH 2.6 (tan). (<b>C</b>) Zoomed view of the differences in VR-IX observed between models of the BuV1 capsid at pH 7.4 (green), 4.0 (teal), and 2.6 (tan).</p>
Full article ">Figure 7
<p>pH-dependent differences in the five-fold pore. Cartoon representation of the pentamers of BuV1 capsid around the 5-fold symmetry axis at (<b>A</b>) pH 7.4, (<b>B</b>) pH 4.0, and (<b>C</b>) pH 2.6. Q157 is shown in stick conformation with the electron density map overlayed at 1.5σ. The 5-fold pore is shown as a circle, defined by the C<span class="html-italic">δ</span> of Q157. The radius from the center is indicated in Å. The red arrow in B represents the 90° shift between the extended and compacted rotamers.</p>
Full article ">Figure 8
<p>Structures of BuV1 yield insight into the viral lifecycle. Diagram showing the proposed viral lifecycle gleaned from the biophysical and structural characterization of BuV1 at different stages of the viral lifecycle. Reconstructed electron density maps are shown at 2σ as follows: Step 1: pH 2.6, Steps 2–4: 6SLN, Step 5a and 5b: pH 4.0, and Step 6–7: pH 7.4. VP1u externalized is postulated due to the alternative conformation of Q157 observed at pH 4.0, which leads to a 5-fold pore with a wider radius.</p>
Full article ">
22 pages, 2085 KiB  
Review
Neuraminidase-1 (NEU1): Biological Roles and Therapeutic Relevance in Human Disease
by Jingxia Du, Hanqi Shui, Rongjun Chen, Yibo Dong, Chengyao Xiao, Yue Hu and Nai-Kei Wong
Curr. Issues Mol. Biol. 2024, 46(8), 8031-8052; https://doi.org/10.3390/cimb46080475 - 26 Jul 2024
Viewed by 511
Abstract
Neuraminidases catalyze the desialylation of cell-surface glycoconjugates and play crucial roles in the development and function of tissues and organs. In both physiological and pathophysiological contexts, neuraminidases mediate diverse biological activities via the catalytic hydrolysis of terminal neuraminic, or sialic acid residues in [...] Read more.
Neuraminidases catalyze the desialylation of cell-surface glycoconjugates and play crucial roles in the development and function of tissues and organs. In both physiological and pathophysiological contexts, neuraminidases mediate diverse biological activities via the catalytic hydrolysis of terminal neuraminic, or sialic acid residues in glycolipid and glycoprotein substrates. The selective modulation of neuraminidase activity constitutes a promising strategy for treating a broad spectrum of human pathologies, including sialidosis and galactosialidosis, neurodegenerative disorders, cancer, cardiovascular diseases, diabetes, and pulmonary disorders. Structurally distinct as a large family of mammalian proteins, neuraminidases (NEU1 through NEU4) possess dissimilar yet overlapping profiles of tissue expression, cellular/subcellular localization, and substrate specificity. NEU1 is well characterized for its lysosomal catabolic functions, with ubiquitous and abundant expression across such tissues as the kidney, pancreas, skeletal muscle, liver, lungs, placenta, and brain. NEU1 also exhibits a broad substrate range on the cell surface, where it plays hitherto underappreciated roles in modulating the structure and function of cellular receptors, providing a basis for it to be a potential drug target in various human diseases. This review seeks to summarize the recent progress in the research on NEU1-associated diseases and highlight the mechanistic implications of NEU1 in disease pathogenesis. An improved understanding of NEU1-associated diseases should help accelerate translational initiatives to develop novel or better therapeutics. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structures and possible modifications of sialic acids [<a href="#B14-cimb-46-00475" class="html-bibr">14</a>,<a href="#B15-cimb-46-00475" class="html-bibr">15</a>,<a href="#B16-cimb-46-00475" class="html-bibr">16</a>,<a href="#B17-cimb-46-00475" class="html-bibr">17</a>].</p>
Full article ">Figure 2
<p>Scheme showing interactions between the components of the NEU1-EBP-PPCA complex.</p>
Full article ">Figure 3
<p>NEU1-regulated signaling pathways implicated in disease pathogenesis across different systems.</p>
Full article ">Figure 4
<p>Etiological involvement of NEU1 dysfunction in diseases across various systems.</p>
Full article ">
14 pages, 1925 KiB  
Review
Antimalarial Drugs at the Intersection of SARS-CoV-2 and Rheumatic Diseases: What Are the Potential Opportunities?
by Saule Abisheva, Kristina Rutskaya-Moroshan, Gulnaz Nuranova, Tansholpan Batyrkhan and Anilim Abisheva
Medicina 2024, 60(7), 1171; https://doi.org/10.3390/medicina60071171 - 19 Jul 2024
Viewed by 572
Abstract
Background and Objectives: The coronavirus disease of 2019 (COVID-19) pandemic has posed a serious threat to humanity and is considered a global health emergency. Antimalarial drugs (ADs) have been used in the treatment of immuno-inflammatory arthritis (IIA) and coronavirus infection (COVID-19). The [...] Read more.
Background and Objectives: The coronavirus disease of 2019 (COVID-19) pandemic has posed a serious threat to humanity and is considered a global health emergency. Antimalarial drugs (ADs) have been used in the treatment of immuno-inflammatory arthritis (IIA) and coronavirus infection (COVID-19). The aim of this review is to analyze the current knowledge about the immunomodulatory and antiviral mechanisms of action, characteristics of use, and side effects of antimalarial drugs. Material and Methods: A literature search was carried out using PubMed, MEDLINE, SCOPUS, and Google Scholar databases. The inclusion criteria were the results of randomized and cohort studies, meta-analyses, systematic reviews, and original full-text manuscripts in the English language containing statistically confirmed conclusions. The exclusion criteria were summary reports, newspaper articles, and personal messages. Qualitative methods were used for theoretical knowledge on antimalarial drug usage in AIRDs and SARS-CoV-2 such as a summarization of the literature and a comparison of the treatment methods. Results: The ADs were considered a “candidate” for the therapy of a new coronavirus infection due to mechanisms of antiviral activity, such as interactions with endocytic pathways, the prevention of glycosylation of the ACE2 receptors, blocking sialic acid receptors, and reducing the manifestations of cytokine storms. The majority of clinical trials suggest no role of antimalarial drugs in COVID-19 treatment or prevention. These circumstances do not allow for their use in the treatment and prevention of COVID-19. Conclusions: The mechanisms of hydroxychloroquine are related to potential cardiotoxic manifestations and demonstrate potential adverse effects when used for COVID-19. Furthermore, the need for high doses in the treatment of viral infections increases the likelihood of gastrointestinal side effects, the prolongation of QT, and retinopathy. Large randomized clinical trials (RCTs) have refuted the fact that there is a positive effect on the course and results of COVID-19. Full article
(This article belongs to the Section Infectious Disease)
Show Figures

Figure 1

Figure 1
<p>Stages of the progression of SARS-CoV-2. Notes: ARDS: acute respiratory distress syndrome; SIRS: systemic inflammatory response syndrome; CRP: C-reactive protein; IL: interleukin; TNF-α: tumor necrosis factor α; GM-CSF: granulocyte-macrophage colony-stimulating factor.</p>
Full article ">Figure 2
<p>The mechanisms of antiviral and immunomodulatory activity ADs in SARS-CoV-2 infection. Notes: ADs: antimalarial drugs, ACE2: angiotensin-converting enzyme 2; IL: interleukin; APCs: antigen-presenting cells; TLRs: Toll-like receptors; MAPK: mitogen-activated protein kinase; ARDS: acute respiratory distress syndrome.</p>
Full article ">Figure 3
<p>Comparison of AD use in the treatment of rheumatic diseases and coronavirus infection. Notes: AIRDs: autoimmune rheumatic diseases; RCTs: randomized clinical trials; HCQ: hydroxychloroquine.</p>
Full article ">
11 pages, 2183 KiB  
Article
Glioma-Associated Sialoglycans Drive the Immune Suppressive Phenotype and Function of Myeloid Cells
by Lenneke A. M. Cornelissen, Kim C. M. Santegoets, Esther D. Kers-Rebel, Sandra A. J. F. H. Bossmann, Mark Ter Laan, Daniel Granado and Gosse J. Adema
Pharmaceutics 2024, 16(7), 953; https://doi.org/10.3390/pharmaceutics16070953 - 19 Jul 2024
Viewed by 604
Abstract
The tumor microenvironment of glioblastoma IDH-wildtype is highly immune suppressive and is characterized by a strong component of myeloid-derived suppressor cells (MDSCs). To interfere with the immune suppressive functions of MDSCs, a comprehensive understanding on how MDSCs acquire their suppressive phenotype is essential. [...] Read more.
The tumor microenvironment of glioblastoma IDH-wildtype is highly immune suppressive and is characterized by a strong component of myeloid-derived suppressor cells (MDSCs). To interfere with the immune suppressive functions of MDSCs, a comprehensive understanding on how MDSCs acquire their suppressive phenotype is essential. Previously, we and others have shown a distinct Sialic acid-binding immunoglobulin-like lectin (Siglec) receptor expression profile for MDSCs in glioblastoma. Siglec receptors can transmit inhibitory signals comparable to PD-1 and are suggested to act as glyco-immune checkpoints. Here, we investigated how glioma specific Siglec-sialic acid interactions influence myeloid immune suppressive functions. Co-culturing monocytes with glioblastoma cells induced CD163 expression on the monocytes. Upon desialylation of the glioblastoma cells, this induction of CD163 was hampered, and furthermore, the monocytes were now able to secrete higher amounts of IL-6 and TNFα compared to fully sialylated glioblastoma cells. Additionally, Siglec-specific triggering using anti-Siglec-7 or Siglec-9 antibodies displayed a decreased TNFα secretion by the monocytes, validating the role of the Siglec–Sialic axis in the co-culture experiments. Together, our results demonstrate that glioblastoma cells induce a myeloid immune-suppressive phenotype that could be partly rescued by lowering the glioblastoma-associated sialic acid levels. This manuscript supports further research of the Siglec–Sialic acid axis in the context of glioblastoma and its potential to improve clinical outcome. Full article
(This article belongs to the Special Issue Novel Therapeutic Strategies for Glioblastoma)
Show Figures

Figure 1

Figure 1
<p>Sialic acid inhibitors stably reduce the sialoglycan content in the human glioblastoma cell line T98G. (<b>A</b>) The two sialic acid inhibitors Ac<sub>5</sub>3F<sub>ax</sub>Neu5Ac and SiaFEt tested cause a dose-dependent inhibition of sialic acid expression on T98G cells compared to DMSO control. T98G cells were treated for 4 days, and expression of α2-3- and α2-6-linked sialic acid was analyzed using MALII and SNA lectins, respectively. The data presents <span class="html-italic">n</span> = 2 independent experiments. (<b>B</b>) T98G cells were treated with either 1000 µM Ac<sub>5</sub>3F<sub>ax</sub>Neu5Ac or 125 µM SiaFEt. After 4 days, the inhibitors were extensively washed away. The T98G cells display a reduction is sialic acid expression for up to 4 days.</p>
Full article ">Figure 2
<p>Sialic acids on T98G glioblastoma cells drive immune-suppressive phenotypes in monocytes. T98G cells were pretreated with sialic acid inhibitors or DMSO controls, and were subsequently co-cultured with monocytes for 4 days. (<b>A</b>,<b>B</b>) Flow cytometry analysis of MHC-II, CD163, CD206, CD86, and Siglec-1 after 4 days of co-culture. Each dot represents one biological replicate. MFI; mean fluorescence intensity. (<b>C</b>) After 4 days, cells were stimulated with 100 ng/mL LPS overnight. Supernatant was collected and cytokine secretion was analyzed. Each dot represents one of the duplo or triplo measurements of one biological replicate. A one-way ANOVA test was performed (** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 3
<p>Sialic acids on T98G glioblastoma cells drive immune-suppressive phenotypes in glioma patient-derived monocytes. T98G cells were pretreated with sialic acid inhibitors or DMSO controls, and were subsequently co-cultured with monocytes obtained from healthy controls or glioma patients in parallel. After 4 days, MHC-II, CD163, CD206, and Siglec-1 expression were analyzed using flow cytometry. Each dot represents one biological replicate.</p>
Full article ">Figure 4
<p>Sialic acids on T98G glioblastoma cells change cytokine secretion profiles in glioma patient and healthy control derived monocytes. T98G cells were pretreated with sialic acid inhibitors or DMSO control, and were subsequently co-cultured with monocytes obtained from healthy controls (HC) or glioma patients (PT) in parallel. After 4 days, cells were stimulated with 100 ng/mL LPS overnight. Supernatant was collected and cytokine secretion was analyzed. Each dot represents one of the duplo or triplo measurements of one biological replicate. A one-way ANOVA test was performed (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 5
<p>Siglec triggering of monocytes results in reduced TNFα secretion. (<b>A</b>) Monocytes (CD14<sup>+</sup> PBMCs) express both Siglec-7 and Siglec-9, which is not present on CD3<sup>+</sup> PBMCs. Each dot represents one biological replicate. (<b>B</b>) Monocytes were stimulated with 10 ng/mL LPS in combination with 5 µg/mL anti-Siglec-7, 5 µg/mL anti-Siglec-9, or 5 µg/mL isotype monoclonal antibodies (mAb). Both Siglec-7 and Siglec-9 mAb stimulation hampered TNFα secretion upon stimulation with LPS. Data are presented as Nested data of <span class="html-italic">n</span> = 9 (Siglec-7) or <span class="html-italic">n</span> = 6 (Siglec-9) biological replicates, with each performed in duplo. A paired <span class="html-italic">t</span>-test was performed. (*** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">
15 pages, 5108 KiB  
Article
Association of Serum Proteases and Acute Phase Factors Levels with Survival Outcomes in Patients with Colorectal Cancer
by Tadeusz Sebzda, Jakub Karwacki, Anna Cichoń, Katarzyna Modrzejewska, Jerzy Heimrath, Mirosław Łątka, Jan Gnus and Jakub Gburek
Cancers 2024, 16(13), 2471; https://doi.org/10.3390/cancers16132471 - 6 Jul 2024
Viewed by 629
Abstract
Colorectal cancer (CRC) represents a substantial burden on global healthcare, contributing to significant morbidity and mortality worldwide. Despite advances in screening methodologies, its incidence remains high, necessitating continued efforts in early detection and treatment. Neoplastic invasion and metastasis are primary determinants of CRC [...] Read more.
Colorectal cancer (CRC) represents a substantial burden on global healthcare, contributing to significant morbidity and mortality worldwide. Despite advances in screening methodologies, its incidence remains high, necessitating continued efforts in early detection and treatment. Neoplastic invasion and metastasis are primary determinants of CRC lethality, emphasizing the urgency of understanding underlying mechanisms to develop effective therapeutic strategies. This study aimed to explore the potential of serum biomarkers in predicting survival outcomes in CRC patients, with a focus on cathepsin B (CB), leukocytic elastase (LE), total sialic acid (TSA), lipid-associated sialic acid (LASA), antitrypsin activity (ATA), C-reactive protein (CRP), and cystatin C (CC). We recruited 185 CRC patients and 35 healthy controls, assessing demographic variables, tumor characteristics, and 7 serum biomarker levels, including (1) CB, (2) LE, (3) TSA, (4) LASA, (5) ATA, (6) CRP, and (7) CC. Statistical analyses included ANOVA with Tukey’s post hoc tests and MANOVA for continuous variables. Student’s t-test was used for dependent samples, while non-parametric tests like Mann–Whitney U and Wilcoxon signed-rank tests were applied for variables deviating from the normal distribution. Categorical variables were assessed using chi-square and Kruskal-Wallis tests. Spearman’s rank correlation coefficient was utilized to examine variable correlations. Survival analysis employed the Kaplan–Meier method with a log-rank test for comparing survival times between groups. Significant associations were observed between CB (p = 0.04), LE (p = 0.01), and TSA (p = 0.008) levels and survival outcomes in CRC patients. Dukes’ classification stages also showed a significant correlation with survival (p = 0.001). However, no significant associations were found for LASA, ATA, CRP, and CC. Multivariate analysis of LE, TSA, and ATA demonstrated a notable correlation with survival (p = 0.041), notwithstanding ATA’s lack of significance in univariate analysis (p = 0.13). CB, LE, and TSA emerged as promising diagnostic markers with prognostic value in CRC, potentially aiding in early diagnosis and treatment planning. Further research is needed to validate these findings and explore additional prognostic indicators. Full article
Show Figures

Figure 1

Figure 1
<p>The survival analysis of patients with colon adenocarcinoma at serum cathepsin B threshold of 11.2248 mU/L. The Kaplan–Meier test was used to perform the survival analysis. <span class="html-italic">p</span>: statistical significance; CB: cathepsin B.</p>
Full article ">Figure 2
<p>The survival analysis of patients with colon adenocarcinoma at serum leukocytic elastase threshold of 534 μg/L. The Kaplan–Meier test was used to perform the survival analysis. <span class="html-italic">p</span>: statistical significance; LE: leukocytic elastase.</p>
Full article ">Figure 3
<p>The survival analysis of patients with colon adenocarcinoma at serum total sialic acid threshold of 75.34 mg%. The Kaplan–Meier test was used to perform the survival analysis. <span class="html-italic">p</span>: statistical significance; TSA: total sialic acid.</p>
Full article ">Figure 4
<p>The survival analysis of patients with colon adenocarcinoma at serum lipid-associated sialic acid threshold of 0.739 mg%. The Kaplan–Meier test was used to perform the survival analysis. <span class="html-italic">p</span>: statistical significance; LASA: lipid-associated sialic acid.</p>
Full article ">Figure 5
<p>The survival analysis of patients with colon adenocarcinoma at serum antitrypsin activity threshold of 2400 U/mL. The Kaplan–Meier test was used to perform the survival analysis. <span class="html-italic">p</span>: statistical significance; ATA: antitrypsin activity.</p>
Full article ">Figure 6
<p>The survival analysis of patients with colon adenocarcinoma at serum C-reactive protein threshold of 47.8 mg/L. The Kaplan–Meier test was used to perform the survival analysis. <span class="html-italic">p</span>: statistical significance; CRP: C-reactive protein.</p>
Full article ">Figure 7
<p>The survival analysis of patients with colon adenocarcinoma at serum cystatin C threshold of 1.55 mg/L. The Kaplan–Meier test was used to perform the survival analysis. <span class="html-italic">p</span>: statistical significance; CC: cystatin C.</p>
Full article ">Figure 8
<p>The survival analysis of patients with colon adenocarcinoma at combined threshold of −0.113 mg/L for mulitparameter model of leukocytic elastase, total sialic acid, and antitrypsin activity. The Kaplan–Meier test was used to perform the survival analysis. <span class="html-italic">p</span>: statistical significance.</p>
Full article ">Figure 9
<p>The graphical summary of the association of investigated biochemical factors and survival outcomes in colorectal adenocarcinoma patients. Our study revealed that decreased levels of cathepsin B and leukocytic elastase, along with elevated levels of total sialic acid, were associated with poorer survival outcomes. However, the association of lipid-associated sialic acid, antitrypsin activity, C-reactive protein, and cystatin C with survival outcomes was found to be non-significant. In multivariate survival analysis, only a combination of leukocytic elastase, total sialic acid, and antitrypsin activity showed a correlation with survival outcomes. <span class="html-italic">p</span>: probability; NS: non-significant.</p>
Full article ">
14 pages, 25616 KiB  
Article
Reduction in Xenogeneic Epitopes on Porcine Endothelial Cells by Periodate Oxidation
by Jonas Thom, Nathalie Roters, Slavica Schuemann, Birgit Andrée, Falk F. R. Buettner, Andres Hilfiker, Tobias Goecke and Robert Ramm
Biomedicines 2024, 12(7), 1470; https://doi.org/10.3390/biomedicines12071470 - 3 Jul 2024
Viewed by 481
Abstract
Background: Patterns of humoral immune responses represent a major hurdle in terms of pig-to-human xenotransplantation approaches. The best-known xenogeneic glycan antigens present in pigs are the αGal (Galili antigen) and the non-human sialic acid Neu5Gc. As there are further differences between porcine and [...] Read more.
Background: Patterns of humoral immune responses represent a major hurdle in terms of pig-to-human xenotransplantation approaches. The best-known xenogeneic glycan antigens present in pigs are the αGal (Galili antigen) and the non-human sialic acid Neu5Gc. As there are further differences between porcine and human cellular surface glycosylation, a much broader range of glycan epitopes with xeno-reactive relevance can be anticipated. Therefore, we set out to chemically modify porcine cellular surface glycans in a global approach by applying sodium periodate (NaIO4) oxidation. Methods: Porcine endothelial cells were exposed to oxidation with 1 to 5 mM NaIO4 for different time periods at 37 °C or 4 °C and under static or dynamic conditions. The impact on cellular survival was determined by applying live/dead assays. Oxidation of αGal-epitopes was assessed by fluorescence microscopy-based quantification of isolectin-B4 (IL-B4) staining. Overall immunogenicity of porcine cells was determined by human serum antibody binding. Results: Treatment of porcine endothelial cells and tissues with NaIO4 led to reduced binding of the αGal-specific IL-B4 and/or human serum antibodies. NaIO4 was revealed to be cytotoxic when performed at elevated temperatures and for a prolonged time. However, by applying 2 mM NaIO4 for 60 min at 4 °C, a high extent of cellular viability and a relevant reduction in detectable αGal epitope were observed. No differences were detected irrespectively on whether the cells were oxidized under static or flow conditions. Conclusions: Glycan epitopes on living cells can be oxidized with NaIO4 while maintaining their viability. Accordingly, this strategy holds promise to prevent immune reactions mediated by preformed anti-glycan antibodies. Full article
Show Figures

Figure 1

Figure 1
<p>Analysis of isolated PECs. (<b>A</b>) Light microscopy revealed a cobblestone-like growth pattern of the isolated cells. (<b>B</b>,<b>C</b>) Immunocytochemistry of the isolated cells using (<b>B</b>) an antibody against VE-Cadherin (red) or (<b>C</b>) the lectin IL-B4 against αGal (red); nuclei were stained with DAPI (blue).</p>
Full article ">Figure 2
<p>Solubility of NaIO<sub>4</sub> at 4 °C in different solvents. NaIO<sub>4</sub> was dissolved in PBS (<b>A</b>), EBM (<b>B</b>), or saline (<b>C</b>) at the indicated concentrations. In PBS, the formation of a white precipitate was observed starting from a concentration of 2 mM NaIO<sub>4</sub> (orange arrows); n = 2.</p>
Full article ">Figure 3
<p>Cell viability is dependent on oxidation temperature. (<b>A</b>) Survival rates of PECs oxidized with 2 mM NaIO<sub>4</sub> for 40 min at 4 °C or 37 °C with respective controls incubated with PBS (n = 3). (<b>B</b>–<b>G</b>) Exemplary pictures of a live/dead assay (green = live; red = dead) of PECs treated at 4 °C (<b>B</b>–<b>D</b>), respectively, and at 37 °C (<b>E</b>–<b>G</b>). (<b>C</b>,<b>F</b>) NaIO<sub>4</sub> oxidation under static conditions was compared to oxidation under dynamic conditions. (<b>D</b>,<b>G</b>) Experiments were performed with cells from three different donor animals. Scale bar 1000 µm in (<b>B</b>,<b>C</b>,<b>E</b>,<b>F</b>) or 100 µm in (<b>D</b>,<b>G</b>). <span class="html-italic">p</span>-values of ≤ 0.01 (**), respectively ≤ 0.0001 (****).</p>
Full article ">Figure 4
<p>Cell survival is dependent on applied concentrations and treatment duration. (<b>A</b>) The cell survival rate at different concentrations of NaIO<sub>4</sub> over time; (n = 3). (<b>B</b>,<b>E</b>,<b>H</b>,<b>K</b>) Live/dead staining of PECs oxidized using 2–5 mM NaIO<sub>4</sub> for 60, 30, 20, or 10 min. (<b>C</b>,<b>F</b>,<b>I</b>,<b>L</b>) IL-B4 staining of PECs treated with the indicated different concentrations of NaIO<sub>4</sub> and duration. (<b>D</b>,<b>G</b>,<b>J</b>,<b>M</b>) Respective controls; PECs treated with PBS for the indicated periods. Scale bars show (<b>B</b>,<b>E</b>,<b>H</b>,<b>K</b>) 1000 µm or (<b>C</b>,<b>D</b>,<b>F</b>,<b>G</b>,<b>I</b>,<b>J</b>,<b>L</b>,<b>M</b>) 200 µm.</p>
Full article ">Figure 5
<p>NaIO<sub>4</sub> oxidation under dynamic conditions. (<b>A</b>,<b>E</b>,<b>I</b>,<b>M</b>) PECs treated with PBS as controls and stained with IL-B4 or human serum. (<b>B</b>,<b>F</b>) Oxidation for 60 min with 2 mM NaIO<sub>4</sub> caused a similar reduction in IL-B4 staining as static conditions (blue = DAPI; red = IL-B4). (<b>C</b>,<b>G</b>) Even stronger reduction was observed using 3 mM NaIO<sub>4</sub> for 60 min. (<b>I</b>–<b>P</b>) Staining of human IgG, IgA, and IgM showed reduced binding of human serum antibodies to PECs oxidized for (<b>J</b>,<b>N</b>) 60 min with 2 mM and a further reduction with (<b>K</b>,<b>O</b>) 3 mM NaIO<sub>4</sub> (blue = DAPI, green = anti-human IgA, IgG, and IgM (Heavy and Light Chain)). (<b>D</b>,<b>H</b>,<b>L</b>,<b>P</b>) Human ECFCs served as negative controls. Experiments were performed in technical triplicates with cells from one donor animal. Scale bar: 100 µm.</p>
Full article ">Figure 6
<p>αGal turnover. PECs stained with IL-B4 (red) directly after oxidation (<b>A</b>) and corresponding untreated controls (<b>B</b>). PECs cultivated for 24 h after oxidation and stained with IL-B4 (<b>C</b>) and respective unprocessed control (<b>D</b>). The extent of areas positively detected by IL-B4 staining increases on levels of unoxidized tissues after 24 h of further incubation. Experiments were performed as biological duplicates with cells from two independent donor animals. Scale bar: 200 µm.</p>
Full article ">Figure 7
<p>NaIO<sub>4</sub> penetrates into native aortic tissue. Cross-sections of treated (<b>A</b>) and untreated (<b>B</b>) aorta were stained with IL-B4. (<b>A</b>) Oxidation with 2 mM NaIO<sub>4</sub> for 40 min at 4 °C led to a distinct reduction in IL-B4 positivity (red) within the intima layer and parts of the media layer. Cellular nuclei were counterstained with DAPI (blue). (<b>B</b>) Complete media layer and even more the intima layer of untreated aorta exhibit IL-B4 staining. This experiment was performed using tissue from only one animal. Scale bar: 200 µm.</p>
Full article ">
13 pages, 2025 KiB  
Article
Swine Influenza Viruses Isolated from 2019 to 2022 in Shandong Province, China, Exemplify the Dominant Genotype
by Yuzhong Zhao, Lebin Han, Haotian Sang, Sidang Liu, Pingping Yang, Yanmeng Hou and Yihong Xiao
Genes 2024, 15(7), 849; https://doi.org/10.3390/genes15070849 - 27 Jun 2024
Viewed by 505
Abstract
Swine influenza viruses (SIVs) have been circulating in swine globally and are potential threats to human health. During the surveillance of SIVs in Shandong Province, China, from 2019 to 2022, 21 reassortant G4 genotype Eurasian avian-like (EA) H1N1 subtypes containing genes from the [...] Read more.
Swine influenza viruses (SIVs) have been circulating in swine globally and are potential threats to human health. During the surveillance of SIVs in Shandong Province, China, from 2019 to 2022, 21 reassortant G4 genotype Eurasian avian-like (EA) H1N1 subtypes containing genes from the EA H1N1 (HA and NA), 2009 pandemic (pdm/09) H1N1 virus (PB2, PB1, PA, NP, and M), and classical swine (CS) H1N1 (NS) lineages were isolated. The analysis of the key functional amino acid sites in the isolated viruses showed that two mutation sites (190D and 225E) that preferentially bind to the human α2-6 sialic acid receptor were found in HA. In PB2, three mutation sites (271A, 590S, and 591R) that may increase mammalian fitness and a mutation site (431M) that increases pathogenicity in mice were found. A typical human signature marker that may promote infection in humans, 357K, was found in NP. The viruses could replicate efficiently in mouse lungs and turbinates, and one of the H1N1 isolates could replicate in mouse kidneys and brains without prior adaption, which indicates that the viruses potentially pose a threat to human health. Histopathological results showed that the isolated viruses caused typical bronchopneumonia and encephalitis in mice. The results indicate that G4 genotype H1N1 has potential transmissibility to humans, and surveillance should be enhanced, which could provide important information for assessing the pandemic potential of the viruses. Full article
(This article belongs to the Special Issue The Diversity and Evolution of the Animal Virome)
Show Figures

Figure 1

Figure 1
<p>Phylogenetic trees of the PB2, PB1, PA, HA, NP, NA, M, and NS genes of H1N1 subtypes. The trees were generated with MEGA 7.0 using neighbor-joining analysis, and the reliabilities of the trees were assessed by bootstrap analysis with 1000 replicates. The isolates in this study are indicated by black circles.</p>
Full article ">Figure 2
<p>The pathogenicity of the isolates in mice. (<b>A</b>). Mouse body weights were monitored daily for 14 days. The values represent the average scores of overall body weight loss compared with the initial body weight ± standard deviation (SD). (<b>B</b>). Viral titers in lungs and nasal turbinates of the infected mice (n = 3) after 3 DPI were determined in 9-to-10-day-old SPF embryonated chicken eggs. (<b>C</b>). Histopathological analysis of lungs and brains. The lungs or brains of the infected mice were fixed with formalin, embedded in paraffin, stained with hematoxylin and eosin, and observed under a microscope at 200× magnification.</p>
Full article ">Figure 2 Cont.
<p>The pathogenicity of the isolates in mice. (<b>A</b>). Mouse body weights were monitored daily for 14 days. The values represent the average scores of overall body weight loss compared with the initial body weight ± standard deviation (SD). (<b>B</b>). Viral titers in lungs and nasal turbinates of the infected mice (n = 3) after 3 DPI were determined in 9-to-10-day-old SPF embryonated chicken eggs. (<b>C</b>). Histopathological analysis of lungs and brains. The lungs or brains of the infected mice were fixed with formalin, embedded in paraffin, stained with hematoxylin and eosin, and observed under a microscope at 200× magnification.</p>
Full article ">
10 pages, 1405 KiB  
Review
Non-Analog Compounds to Sialic Acid as Inhibitors of Influenza Virus Neuraminidase: An Underexplored Approach for Novel Antivirals―Systematic Review
by Luis Márquez-Domínguez, Carolina Jasso-Miranda, Virginia Sedeño-Monge and Gerardo Santos-López
Sci. Pharm. 2024, 92(2), 33; https://doi.org/10.3390/scipharm92020033 - 19 Jun 2024
Viewed by 1008
Abstract
Influenza poses a significant threat to public health worldwide, particularly among vulnerable populations such as children, the elderly, immunocompromised individuals, and those with chronic diseases. It is associated with high mortality and morbidity rates. Neuraminidase inhibitors play a crucial role in influenza treatment [...] Read more.
Influenza poses a significant threat to public health worldwide, particularly among vulnerable populations such as children, the elderly, immunocompromised individuals, and those with chronic diseases. It is associated with high mortality and morbidity rates. Neuraminidase inhibitors play a crucial role in influenza treatment by mitigating the risk of complications and death. However, the genetic variability of the influenza virus enables the emergence of drug-resistant mutations. This review focuses on the search for new compounds that are not analogous to sialic acid, aiming to inhibit the activity of viral neuraminidase in vitro, viral replication in cell cultures, or animal models. Influenza virus strains that have been reported in the literature present specific mutations that generate resistance to neuraminidase inhibitors. Since these inhibitors bear structural resemblance to sialic acid, the predominant location for these mutations is the enzyme’s active site. Consequently, exploring alternative compound classes becomes imperative to circumvent this interaction pattern. These compounds will introduce diverse molecular frameworks, serving as foundational structures for further development through rational drug design, thereby engendering novel antiviral agents targeting influenza. The potential prospects for developing novel influenza antivirals based on these findings are discussed. Full article
(This article belongs to the Topic Challenges and Opportunities in Drug Delivery Research)
Show Figures

Figure 1

Figure 1
<p>Flow diagram of literature search and study selection.</p>
Full article ">Figure 2
<p>Heat map of influenza virus inhibitors reported in 31 selected articles. The numbers on the <span class="html-italic">x</span>-axis correspond to the numeration of all compounds, which can be found in <a href="#app1-scipharm-92-00033" class="html-app">Supplementary Table S1</a>. It is important to note that some compounds may appear more than once on the heat map, depending on whether they were tested with different viral subtypes or if the same compound was tested in different studies. IC<sub>50</sub>: half maximal inhibitory concentration, CC<sub>50</sub>: half maximal cytotoxic concentration, EC<sub>50</sub>: half maximal effective concentration.</p>
Full article ">Figure 3
<p>Molecular structures of selected compounds. (<b>a</b>) zanamivir, a commercial antiviral; (<b>b</b>,<b>c</b>) compounds selected for their high inhibitory capacity in vitro (IC<sub>50</sub> &lt; 1 μM); (<b>d</b>–<b>f</b>) compounds selected for their inhibitory activity against viral replication (EC<sub>50</sub> &lt; 0.1 μM); (<b>g</b>) compound select for in vivo studies.</p>
Full article ">
22 pages, 8423 KiB  
Article
Artificial and Natural Sweeteners Biased T1R2/T1R3 Taste Receptors Transactivate Glycosylated Receptors on Cancer Cells to Induce Epithelial–Mesenchymal Transition of Metastatic Phenotype
by Elizabeth Skapinker, Rashelle Aldbai, Emilyn Aucoin, Elizabeth Clarke, Mira Clark, Daniella Ghokasian, Haley Kombargi, Merlin J. Abraham, Yunfan Li, David A. Bunsick, Leili Baghaie and Myron R. Szewczuk
Nutrients 2024, 16(12), 1840; https://doi.org/10.3390/nu16121840 - 12 Jun 2024
Viewed by 1164
Abstract
Understanding the role of biased taste T1R2/T1R3 G protein-coupled receptors (GPCR) agonists on glycosylated receptor signaling may provide insights into the opposing effects mediated by artificial and natural sweeteners, particularly in cancer and metastasis. Sweetener-taste GPCRs can be activated by several active states [...] Read more.
Understanding the role of biased taste T1R2/T1R3 G protein-coupled receptors (GPCR) agonists on glycosylated receptor signaling may provide insights into the opposing effects mediated by artificial and natural sweeteners, particularly in cancer and metastasis. Sweetener-taste GPCRs can be activated by several active states involving either biased agonism, functional selectivity, or ligand-directed signaling. However, there are increasing arrays of sweetener ligands with different degrees of allosteric biased modulation that can vary dramatically in binding- and signaling-specific manners. Here, emerging evidence proposes the involvement of taste GPCRs in a biased GPCR signaling crosstalk involving matrix metalloproteinase-9 (MMP-9) and neuraminidase-1 (Neu-1) activating glycosylated receptors by modifying sialic acids. The findings revealed that most natural and artificial sweeteners significantly activate Neu-1 sialidase in a dose-dependent fashion in RAW-Blue and PANC-1 cells. To confirm this biased GPCR signaling crosstalk, BIM-23127 (neuromedin B receptor inhibitor, MMP-9i (specific MMP-9 inhibitor), and oseltamivir phosphate (specific Neu-1 inhibitor) significantly block sweetener agonist-induced Neu-1 sialidase activity. To assess the effect of artificial and natural sweeteners on the key survival pathways critical for pancreatic cancer progression, we analyzed the expression of epithelial-mesenchymal markers, CD24, ADLH-1, E-cadherin, and N-cadherin in PANC-1 cells, and assess the cellular migration invasiveness in a scratch wound closure assay, and the tunneling nanotubes (TNTs) in staging the migratory intercellular communication. The artificial and natural sweeteners induced metastatic phenotype of PANC-1 pancreatic cancer cells to promote migratory intercellular communication and invasion. The sweeteners also induced the downstream NFκB activation using the secretory alkaline phosphatase (SEAP) assay. These findings elucidate a novel taste T1R2/T1R3 GPCR functional selectivity of a signaling platform in which sweeteners activate downstream signaling, contributing to tumorigenesis and metastasis via a proposed NFκB-induced epigenetic reprogramming modeling. Full article
(This article belongs to the Special Issue Effects of Sugars and Sugar Alternatives on Human Health and Disease)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Molecular signaling platform on the cell surface reveals an oligomeric selectivity of T1R2/T1R3 GPCR-biased functional heteromers with NMBR to induce the TLR activation signaling axis. Citation: Taken in part from Abdulkhalek et al. [<a href="#B6-nutrients-16-01840" class="html-bibr">6</a>] Cellular signalling 2012, 24, 2035–2042. Publisher and licensee Elsevier. This is an Open Access article which permits unrestricted non-commercial use, provided the original work is properly cited. (<b>B</b>,<b>C</b>) Sialidase activity is associated with artificial sweeteners treatment of live RAW-blue macrophage cells. The phase contrast image of the cells represents the cell numbers used in the sialidase assay.</p>
Full article ">Figure 2
<p>(<b>A</b>–<b>C</b>) Sialidase activity is associated with natural sweeteners glucose, stevia and monk using live RAW-blue macrophage cells.</p>
Full article ">Figure 3
<p>(<b>A</b>) Molecular signaling platform reveals an oligomeric selectivity of T1R2/T1R3 GPCR-biased functional heteromers with NMBR to induce the RTK activation signaling axis. This signaling platform potentiates MMP-9 and Neu-1 crosstalk on the cell surface, which is essential for activating RTK. Citation: Taken in part from DuBois [<a href="#B12-nutrients-16-01840" class="html-bibr">12</a>] Physiology &amp; Behavior 2016, 164, 453–463, Publisher and licensee Elsevier. and Liauchonak et al. [<a href="#B4-nutrients-16-01840" class="html-bibr">4</a>] Nutrients 2019, 11, 644, Publisher and licensee MDPI. This is an Open Access article which permits unrestricted non-commercial use, provided the original work is properly cited. (<b>B</b>) Sialidase activity is associated with natural and artificial sweeteners treatment of live pancreatic PANC-1 cancer cells.</p>
Full article ">Figure 4
<p>(<b>A</b>) Sweetener T1R2/T1R3 GPCR agonists activate NMBR GPCR tethered to TLR4 and MMP-9 to induce Neu-1 sialidase in macrophage cells. Citation: Taken in part from Abdulkhalek et al. [<a href="#B6-nutrients-16-01840" class="html-bibr">6</a>] Cellular signalling 2012, 24, 2035–2042. Publisher and licensee Elsevier. This is an Open Access article which permits unrestricted non-commercial use, provided the original work is properly cited. (<b>B</b>) Artificial and natural sweeteners induce NFκB-Dependent SEAP activity in live RawBlue macrophage cells.</p>
Full article ">Figure 5
<p>Analyses of pancreatic PANC-1 cancer cells for (<b>A</b>) CD24 and (<b>B</b>) ALDH1A1 stem-like markers and (<b>C</b>) E-and (<b>D</b>) N-cadherin EMT markers following 24 h exposure to saccharin, acesulfame, advantame and glucose using immunocytochemistry analyses. ns = non-significant.</p>
Full article ">Figure 6
<p>Half maximal effective concentration (EC50) of artificial sweeteners (<b>A</b>,<b>B</b>) saccharin, (<b>C</b>,<b>D</b>) acesulfame K and (<b>E</b>,<b>F</b>) advantame to induce an invasive epithelial-mesenchymal transition (EMT) N-cadherin expression following 22 h exposure using immunocytochemistry analyses.</p>
Full article ">Figure 7
<p>Half maximal effective concentration (EC50) of natural sweeteners (<b>A</b>,<b>B</b>) lactose, (<b>C</b>,<b>D</b>) monk and (<b>E</b>,<b>F</b>) stevia to induce an invasive epithelial-mesenchymal transition (EMT) N-cadherin expression following 24 h exposure using immunocytochemistry analyses.</p>
Full article ">Figure 8
<p>(<b>A</b>) Structures of lactose, galactose, and glucose. (<b>B</b>) Natural sweeteners lactose, and not glucose and galactose, induce an invasive epithelial-mesenchymal transition (EMT) N-cadherin expression following 24-h exposure using immunocytochemistry analyses.</p>
Full article ">Figure 9
<p>Natural (stevia and lactose) and artificial (advantame and neotame) sweeteners enhance the migratory potential of PANC-1 cancer cells using the scratch wound assay. The wound closure rate of untreated PANC-1 cells was over 24 h. The best-fit slope represents the rate in mm/hr of wound closure.</p>
Full article ">Figure 10
<p>Neotame, lactose and stevia sweeteners induce tunneling nanotubes (TNT) in PANC-1 cells.</p>
Full article ">Figure 11
<p>Dimeric taste receptor T1R2/T1R3 participates in a multimeric receptor complex with neuromedin B receptor (NMBR), neuraminidase 1 (Neu-1), and Toll-like receptor (TLR). Here, the biased functional T1R2/T1R3 GPCR oligomeric platform potentiates Neu-1 and MMP-9 cell surface crosstalk, mediating TLR glycosylation modification and transactivation and subsequent NF-κB-induced epigenetic rewiring. Artificial sweeteners binding to T1R2/T1R3 GPCR are illustrated. Notes: Sweetener stimulation of the biased T1R2/T1R3 GPCR induces receptor heterodimerization with NMBR, wherein NMBR-induced activation of SNAIL to induce MMP-9 endopeptidase to cleave the elastin binding protein (EBP) and expose the catalytic sialidase domain of Neu-1. The sialidase domain of Neu-1 hydrolyzes α-2,3-sialic acid from the glycosylated TLR receptor, reducing the steric hindrance and facilitating TLR dimerization, activation, and downstream cellular signaling. The resultant downstream signaling mediates the phosphorylation of the IkB subunit, which facilitates the translocation of NF-κB to the nucleus, enabling epigenetic modulation of gene expression [<a href="#B28-nutrients-16-01840" class="html-bibr">28</a>,<a href="#B44-nutrients-16-01840" class="html-bibr">44</a>]. Citation: Reprinted/Adapted with permission Jakowiecki et al. [<a href="#B45-nutrients-16-01840" class="html-bibr">45</a>] Molecules 2021, 26, 2456, Publisher and licensee MDPI, Reber et al. [<a href="#B43-nutrients-16-01840" class="html-bibr">43</a>] PLoS One 2009, 4, e4393, Publisher and licensee PLOS and Marquardt et al. [<a href="#B46-nutrients-16-01840" class="html-bibr">46</a>] J Hepatol 2010, 53, 568-577, Publisher and licensee Elsevier. These articles are open-access articles distributed under the terms and conditions of the Creative Commons Attribution (CCBY) license (<a href="http://creativecommons.org/licenses/by/4.0/" target="_blank">http://creativecommons.org/licenses/by/4.0/</a> (accessed on 23 April 2021)), It permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.</p>
Full article ">
18 pages, 3022 KiB  
Article
Neutrophil Depletion Changes the N-Glycosylation Pattern of IgG in Experimental Murine Sepsis
by Kursat O. Yaykasli, Karin A. van Schie, René E. M. Toes, Manfred Wuhrer, Carolien A. M. Koeleman, Galyna Bila, Nazar Negrych, Georg Schett, Jasmin Knopf, Martin Herrmann and Rostyslav Bilyy
Int. J. Mol. Sci. 2024, 25(12), 6478; https://doi.org/10.3390/ijms25126478 - 12 Jun 2024
Viewed by 856
Abstract
Sepsis is a life-threatening condition with a rising disease burden worldwide. It is a multifactorial disease and is defined as a dysregulated host response to infection. Neutrophils have been shown to be involved in the pathogenesis of sepsis by exacerbating inflammation. However, the [...] Read more.
Sepsis is a life-threatening condition with a rising disease burden worldwide. It is a multifactorial disease and is defined as a dysregulated host response to infection. Neutrophils have been shown to be involved in the pathogenesis of sepsis by exacerbating inflammation. However, the exact effector mechanism of action still remains a mystery. Changes in the glycosylation pattern of the immunoglobulin G (IgG) Fc region are described for several diseases including meningococcal sepsis. In this study, we investigated the possible contribution of neutrophils and neutrophil implication, potentially related to degranulation or neutrophil extracellular trap (NET) formation in changing the IgG Fc N-glycosylation pattern in a murine sepsis model. We have measured the serum level of cytokines/chemokines and immunoglobulins, the serum activity of neutrophil elastase (NE), and analyzed the IgG Fc glycosylation pattern by Liquid Chromatography-Electrospray Ionization-Mass Spectrometry (LC-ESI-MS) and Lectin enzyme-linked immunosorbent assay (ELISA). We observed an increased activity of NE- and neutrophil-associated cytokines such as keratinocyte chemoattractant (KC) with the development of sepsis. Regarding the IgG Fc N-glycosylation, we observed an increase in fucosylation and α1,3-galactosylation and a decrease for sialyation. Interestingly, these changes were not uniform for all IgG subclasses. After depletion of neutrophils, we saw a change in the exposure of fucose and α2,6-linked sialic acid during the time course of our experimental sepsis model. In conclusion, neutrophils can influence changes in the IgG glycosylation pattern in experimental sepsis. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Immunoglobulin-associated glycans change within the course of experimental murine sepsis in C57/BL6N mice. Time course of the levels of IgG (<b>A</b>) and IgM (<b>B</b>) before and after sepsis induction is displayed. The serum immunoglobulin-associated glycans were detected by the lectins AAL (core α1,6-fucose) (<b>C</b>,<b>D</b>), LCA (fucosylated trimannose) (<b>E</b>,<b>F</b>), SNA (terminal a2,6-sialic acid) (<b>G</b>,<b>H</b>), and PSqL (terminal a 2,6-sialic acid of N-glycans) (<b>I</b>,<b>J</b>). The levels of lectins were normalized to the levels of IgG and IgM. Note that the glycosylation of both IgG and IgM changed substantially in the course of sepsis. AAL = Aleuria aurantia lectin; LCA = Lens culinaris agglutinin; SNA = Sambucus nigra agglutinin; pSqL = Polyporus squamosus lectin. Kruskall-Wallis test with Dunn’s multiple comparisons post-test was used to compare differences among time points (n = 12). * <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, •—represents individual data points.</p>
Full article ">Figure 2
<p>Changes in the composition of the IgG Fc glycans over the time course in experimental sepsis in the serum of C57/BL6N mice. Glycopeptide composition of the Fc region of IgG1 (<b>A</b>–<b>H</b>), IgG2 (<b>I</b>–<b>P</b>), and IgG3 (<b>Q</b>–<b>X</b>) was analyzed by Liquid Chromatography-Electrospray Ionization-Mass Spectrometry (LC-ESI-MS). The relative percentages of complex monoantennary and diantennary glycans of IgG Fc and the glycosylation traits: (I) ß1,4-galactosylation, (II) ratio of ß1,4-galactosylation and α2,6 sialylation, (III) α1,3-galactosylation, (IV) α2,6 sialylation, (V) fucosylation, and (VI) hybrid for diantennary glycans were calculated. Kruskall-Wallis test with Dunn’s multiple comparisons post-test was used to compare differences among time points (n = 12). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, •—represents individual data points.</p>
Full article ">Figure 3
<p>Changes in the serum immunoglobulin levels over the time course of experimental sepsis in C57/BL6N mice. Serum immunoglobulins concentrations were measured by LEGENDplex™ bead assay before and after sepsis induction in C57/BL6N mice (only significant data are shown, namely for IgG2b (<b>A</b>), IgG3 (<b>B</b>) and IgM (<b>C</b>)). Kruskall-Wallis test with Dunn’s multiple post-test was used to compare differences among time points (n = 12), •—represents individual data points.</p>
Full article ">Figure 4
<p>Changes in neutrophil elastase activity and serum levels of cytokines/chemokines in a model of experimental murine sepsis. (<b>A</b>–<b>E</b>) Sera levels of IFN-β, IFN-y, IL-10, IL-17A, IL-1β, IL-6, IP-10, KC, LIX, MCP-1, MIG, MIP-1a, and TNF-a were measured by a custom-made LEGENDplex™ cytokine/chemokine detection assay before and after sepsis induction in C57/BL6N mice. (<b>F</b>) Changes in the mean fluorescence intensity (MFI) levels of substrate converted by neutrophil elastase (NE) in the sera of C57/BL6N mice during the course of experimental sepsis over time in days. Kruskall-Wallis test with Dunn’s multiple comparisons post-test was used to compare differences among time points (n = 12). Only significant data of the cytokine/chemokine assay are shown, •—represents individual data points.</p>
Full article ">Figure 5
<p>Depletion of neutrophils changed the glycosylation of IgG and IgM bound to IgG. Neutrophils were depleted using the 1A8 depletion antibody (n = 5) or an isotype control (n = 5) during the time course of experimental sepsis in C57/BL6N mice. Next to the serum levels of IgG and IgM bound to IgG, the exposure of the IgG glycans was assessed by lectin ELISA employing the following lectins: (I) AAL (α1,6-linked fucose), (II) LCA (α-linked mannose), and (III) SNA (α2,6-linked sialic acid). The levels were normalized to IgG (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) and IgM (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>). Two-way ANOVA (or Mixed-effects analysis) with Sidak multiple comparisons post-test was used to compare differences among time points.</p>
Full article ">Figure 6
<p>Schematic representation of all glycoforms we studied. The forms with terminal galactose are labeled in red. Please note that all forms bearing sialic acid also contain subterminal galactose residue. N-acetylglucosamine (blue rectangle), fucose (red triangle), mannose (green circle), galactose (yellow circle), sialic acid (light blue rhombus).</p>
Full article ">
19 pages, 4281 KiB  
Article
NMR Studies of the Interactions between Sialyllactoses and the Polysialytransferase Domain for Polysialylation Inhibition
by Bo Lu, Si-Ming Liao, Shi-Jie Liang, Jian-Xiu Li, Xue-Hui Liu, Ri-Bo Huang and Guo-Ping Zhou
Curr. Issues Mol. Biol. 2024, 46(6), 5682-5700; https://doi.org/10.3390/cimb46060340 - 7 Jun 2024
Viewed by 812
Abstract
It is known that sialyllactose (SL) in mammalians is a major source of sialic acid (Sia), which can further form cytidine monophosphate sialic acid (CMP-Sia), and the final product is polysialic acid (polySia) using polysialyltransferases (polySTs) on the neural cell adhesion molecule (NCAM). [...] Read more.
It is known that sialyllactose (SL) in mammalians is a major source of sialic acid (Sia), which can further form cytidine monophosphate sialic acid (CMP-Sia), and the final product is polysialic acid (polySia) using polysialyltransferases (polySTs) on the neural cell adhesion molecule (NCAM). This process is called NCAM polysialylation. The overexpression of polysialylation is strongly related to cancer cell migration, invasion, and metastasis. In order to inhibit the overexpression of polysialylation, in this study, SL was selected as an inhibitor to test whether polysialylation could be inhibited. Our results suggest that the interactions between the polysialyltransferase domain (PSTD) in polyST and CMP-Siaand the PSTD and polySia could be inhibited when the 3′-sialyllactose (3′-SL) or 6′-sialyllactose (6′-SL) concentration is about 0.5 mM or 6′-SL and 3 mM, respectively. The results also show that SLs (particularly for 3′-SL) are the ideal inhibitors compared with another two inhibitors, low-molecular-weight heparin (LMWH) and cytidine monophosphate (CMP), because 3’-SL can not only be used to inhibit NCAM polysialylation, but is also one of the best supplements for infant formula and the gut health system. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

Figure 1
<p>CD spectra of the PSTD alone (black), and in the presence of polySia/CMP-Sia (red), or in the presence of polySia/CMP-Sia/3′-SL (green) (<b>a</b>); CD spectra of the PSTD peptide in the absence of any ligand black and in the presence of polySia/CMP-Sia (red), or in the presence of polySia/CMP-Sia/6′-SL (green) (<b>b</b>).</p>
Full article ">Figure 2
<p>The overlaid <sup>1</sup>H-<sup>15</sup>N HSQC spectra of the PSTD in the absence and presence of 0.5 mM 3′-SL (<b>a</b>), and 1 mM 3′-SL (<b>b</b>), respectively. The obvious changes in chemical shift are residues located in the N-terminus and C-terminal, comparing (<b>a</b>) with (<b>b</b>).</p>
Full article ">Figure 3
<p>CSPs of the PSTD for the PSTD-(CMP-1 mM Sia), the PSTD-(0.5 mM 3′-SL), the PSTD-(1 mM 3′-SL), and the PSTD-(3 mM 3′-SL) interactions, respectively (<b>a</b>); the CSPs of the PSTD for the PSTD-(0.1 mM polySia or PSA), the PSTD-(0.5 mM 3′-SL) and the PSTD-(2 mM 3′-SL) interactions, respectively (<b>b</b>).</p>
Full article ">Figure 3 Cont.
<p>CSPs of the PSTD for the PSTD-(CMP-1 mM Sia), the PSTD-(0.5 mM 3′-SL), the PSTD-(1 mM 3′-SL), and the PSTD-(3 mM 3′-SL) interactions, respectively (<b>a</b>); the CSPs of the PSTD for the PSTD-(0.1 mM polySia or PSA), the PSTD-(0.5 mM 3′-SL) and the PSTD-(2 mM 3′-SL) interactions, respectively (<b>b</b>).</p>
Full article ">Figure 4
<p>The binding ranges of the three ligands, CMP-Sia, 3′-SL and 6′-SL on the PSTD based on the results shown in <a href="#cimb-46-00340-f002" class="html-fig">Figure 2</a>, <a href="#cimb-46-00340-f003" class="html-fig">Figure 3</a>, <a href="#cimb-46-00340-f005" class="html-fig">Figure 5</a> and <a href="#cimb-46-00340-f006" class="html-fig">Figure 6</a>, respectively.</p>
Full article ">Figure 5
<p>The overlaid <sup>1</sup>H-<sup>15</sup>N HSQC spectra of the PSTD in the absence and presence of 1.0 mM 6′-SL (<b>a</b>), and 2.0 mM 6′-SL (<b>b</b>), respectively. The obvious changes in chemical shift are residues K246, R252, T253 in the N-terminus, comparing (<b>a</b>) and (<b>b</b>).</p>
Full article ">Figure 6
<p>The chemical shift perturbations (CSPs) of the PSTD in the presence of 0.5, 1.0, 2.0, and 3.0 mM 6′-SL (<b>a</b>); the CSPs of the PSTD when interacting with 1 mM CMP-Sia, and 1.0 mM 6-SL, respectively (<b>b</b>); and the CSPs of the PSTD when interacting with 0.1 mM PSA, 2.0 mM and 3 mM 6′-SL, respectively (<b>c</b>).</p>
Full article ">Figure 7
<p>Comparison of the CSPs in the PSTD when PSTD interacted with different ligands. The CSPs of the PSTD for the PSTD-0.5 mM 3′-SL, the PSTD-1 mM 6′-SL, the PSTD-1 mM (CMP-Sia), and the PSTD-80 μM LMWH interactions (<b>a</b>); CSPs of the PSTD for the PSTD-0.5 mM 3′-SL, the PSTD-3 mM 6′-SL, the PSTD-80 μM LMWH, and the PSTD-0.1 mM polySia interactions (<b>b</b>).</p>
Full article ">Figure 7 Cont.
<p>Comparison of the CSPs in the PSTD when PSTD interacted with different ligands. The CSPs of the PSTD for the PSTD-0.5 mM 3′-SL, the PSTD-1 mM 6′-SL, the PSTD-1 mM (CMP-Sia), and the PSTD-80 μM LMWH interactions (<b>a</b>); CSPs of the PSTD for the PSTD-0.5 mM 3′-SL, the PSTD-3 mM 6′-SL, the PSTD-80 μM LMWH, and the PSTD-0.1 mM polySia interactions (<b>b</b>).</p>
Full article ">Figure 8
<p>Comparison of the inhibitions of the SL CMP for the interaction between the PSTD and CMP-Sia, and the interaction between the PSTD and polySia. The CSPs of the PSTD for the PSTD-(0.5 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(1 mM CMP-Sia) interactions (<b>a</b>); CSPs of the PSTD for the PSTD-(0.5 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(0.1 mM polySia) interactions (<b>b</b>); CSPs of the PSTD for the PSTD-(1 mM 6′-SL), the PSTD-1 mM CMP, and the PSTD-(1 mM CMP-Sia) interactions (<b>c</b>); CSPs of the PSTD for the PSTD-(3 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(0.1 mM polySia) interactions (<b>d</b>).</p>
Full article ">Figure 8 Cont.
<p>Comparison of the inhibitions of the SL CMP for the interaction between the PSTD and CMP-Sia, and the interaction between the PSTD and polySia. The CSPs of the PSTD for the PSTD-(0.5 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(1 mM CMP-Sia) interactions (<b>a</b>); CSPs of the PSTD for the PSTD-(0.5 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(0.1 mM polySia) interactions (<b>b</b>); CSPs of the PSTD for the PSTD-(1 mM 6′-SL), the PSTD-1 mM CMP, and the PSTD-(1 mM CMP-Sia) interactions (<b>c</b>); CSPs of the PSTD for the PSTD-(3 mM 3′-SL), the PSTD-1 mM CMP, and the PSTD-(0.1 mM polySia) interactions (<b>d</b>).</p>
Full article ">
20 pages, 3184 KiB  
Article
The Dual-Pseudotyped Lentiviral Vector with VSV-G and Sendai Virus HN Enhances Infection Efficiency through the Synergistic Effect of the Envelope Proteins
by Bat-Erdene Jargalsaikhan, Masanaga Muto, Youngeun Been, Shoma Matsumoto, Eiichi Okamura, Tadanobu Takahashi, Yutaka Narimichi, Yuuki Kurebayashi, Hideyuki Takeuchi, Takashi Shinohara, Ryo Yamamoto and Masatsugu Ema
Viruses 2024, 16(6), 827; https://doi.org/10.3390/v16060827 - 23 May 2024
Viewed by 1646
Abstract
A gene delivery system utilizing lentiviral vectors (LVs) requires high transduction efficiency for successful application in human gene therapy. Pseudotyping allows viral tropism to be expanded, widening the usage of LVs. While vesicular stomatitis virus G (VSV-G) single-pseudotyped LVs are commonly used, dual-pseudotyping [...] Read more.
A gene delivery system utilizing lentiviral vectors (LVs) requires high transduction efficiency for successful application in human gene therapy. Pseudotyping allows viral tropism to be expanded, widening the usage of LVs. While vesicular stomatitis virus G (VSV-G) single-pseudotyped LVs are commonly used, dual-pseudotyping is less frequently employed because of its increased complexity. In this study, we examined the potential of phenotypically mixed heterologous dual-pseudotyped LVs with VSV-G and Sendai virus hemagglutinin-neuraminidase (SeV-HN) glycoproteins, termed V/HN-LV. Our findings demonstrated the significantly improved transduction efficiency of V/HN-LV in various cell lines of mice, cynomolgus monkeys, and humans compared with LV pseudotyped with VSV-G alone. Notably, V/HN-LV showed higher transduction efficiency in human cells, including hematopoietic stem cells. The efficient incorporation of wild-type SeV-HN into V/HN-LV depended on VSV-G. SeV-HN removed sialic acid from VSV-G, and the desialylation of VSV-G increased V/HN-LV infectivity. Furthermore, V/HN-LV acquired the ability to recognize sialic acid, particularly N-acetylneuraminic acid on the host cell, enhancing LV infectivity. Overall, VSV-G and SeV-HN synergistically improve LV transduction efficiency and broaden its tropism, indicating their potential use in gene delivery. Full article
(This article belongs to the Section General Virology)
Show Figures

Figure 1

Figure 1
<p>Increased transduction efficiency of LVs pseudotyped with VSV-G and SeV-HN in HEK293FT cells. (<b>A</b>) Schematic diagram of LV pseudotyping with the VSV-G and SeV envelope proteins. Seven types of LV particles were produced using transfer, packaging, Rev, and envelope plasmids in a 4:2:1:1 ratio, as detailed in <a href="#app1-viruses-16-00827" class="html-app">Table S1</a>. (<b>B</b>) The biological and physical titers of LVs. The bar graphs represent TU/mL, and the triangles represent PP/mL. The ratio of TU to PP is shown above each set of LV titers. (<b>C</b>) The expression level of GFP was observed by fluorescence microscopy 48 h after LV transduction at 200 PP/cell in HEK293FT cells. Scale bar, 100 µm. (<b>D</b>) HEK293FT cells were infected with LV particles at the indicated PP/cell and the percentage of GFP-positive cells was determined at 2 days post-infection by flow cytometry. (<b>E</b>) Ultracentrifuged LV particles were subjected to western blotting using VSV-G, whole SeV, and HIV1 p24 antibodies. Data are expressed as the mean ± SD with technical replicates (n = 3). Ordinary one-way ANOVA with Tukey post-hoc test was conducted for (<b>B</b>) and ordinary two-way ANOVA with Tukey post-hoc test was conducted for (<b>D</b>). **** <span class="html-italic">p</span> &lt; 0.0001; ns, not significant; nd, not detected.</p>
Full article ">Figure 2
<p>Viral tropism of LVs with VSV-G and SeV envelope proteins. (<b>A</b>) LV particles used in the tropism assay were produced using transfer, packaging, VSV-G/Rev, SeV-F, and/or SeV-HN envelope plasmids in a 2:1:1:1 ratio, as detailed in <a href="#app1-viruses-16-00827" class="html-app">Table S2</a>. (<b>B</b>) The biological and physical titers of LVs. The bar graphs represent TU/mL, and the triangles represent PP/mL. The ratio of TU to PP is shown above each set of LV titers. (<b>C</b>–<b>H</b>) Human cells: (<b>C</b>) human naïve embryonic stem cells (ESCs), (<b>D</b>) human primary dermal fibroblast cells, (<b>E</b>) colorectal adenocarcinoma cell line Caco-2, (<b>F</b>) human cervical carcinoma derived cell line HeLa, (<b>G</b>) human trophoblast stem cells (TSCs), and (<b>H</b>) human hepatocellular carcinoma cell line Hep3B; (<b>I</b>,<b>J</b>) monkey cells: (<b>I</b>) cynomolgus monkey primary dermal fibroblast cells and (<b>J</b>) African green monkey kidney fibroblast-like cell line COS-7; and (<b>K</b>,<b>L</b>) mouse cells: (<b>K</b>) mouse embryonic stem cells (ESCs) and (<b>L</b>) mouse embryonic fibroblast cell line NIH3T3, were transduced with four types of LV particles at the indicated PP/cell. The infection efficiency was measured 2–4 days post-infection by counting GFP-positive cells via flow cytometry. Data are expressed as the mean ± SD with technical replicates (n = 3). Ordinary one-way ANOVA with Tukey post-hoc test was conducted for (<b>B</b>) and ordinary two-way ANOVA with Tukey post-hoc test was conducted for (<b>C</b>–<b>L</b>). **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, and * <span class="html-italic">p</span> &lt; 0.05; ns, not significant.</p>
Full article ">Figure 3
<p>Relationship between the infection efficiency and the amount of SeV-HN protein in the V/HN-LV particles. (<b>A</b>) Schematic diagram of the production of LV particles using the modified SeV-HN-expressing plasmids. A 2:1:1 ratio of transfer, packaging, and VSV-G/Rev plasmids, and variable amounts of SeV-HN plasmids (unmodified HN, Kozak HN, or Kozak human codon-optimized HN), were used for the optimization of dual-pseudotyped LV particles. (<b>B</b>) The biological and physical titers of LVs. The bar graphs represent TU/mL, and the triangles represent PP/mL. The ratio of TU to PP is shown above each set of LV titers. (<b>C</b>) HEK293FT cells were infected with LV particles at 200 PP/cell, and the infection efficiency was determined by flow cytometry 2 days after transduction. (<b>D</b>) Ultracentrifuged LV particles were subjected to western blotting using VSV-G, whole SeV, and HIV1 p24 antibodies. Data are expressed as the mean ± SD with technical replicates (n = 3) and ordinary one-way ANOVA with Tukey post-hoc test. **** <span class="html-italic">p</span> &lt; 0.0001; ns, not significant.</p>
Full article ">Figure 4
<p>V/HN-LV recognizes the sialylated receptor on the host cell membrane. (<b>A</b>) Schematic diagram showing investigation of the importance of sialic acid on the host cell membrane during V/HN-LV infection using (<b>B</b>) a sialyltransferase inhibitor (STi) and (<b>C</b>) sialidase A treatment. The transduction efficiency of LV particles was determined by counting GFP-positive cells using flow cytometry 48 h after infection at 800 PP/cell. (<b>D</b>) Phase contrast and GFP fluorescence images 2 days after infection in the experimental groups. (<b>E</b>) Schematic illustration shows the glycan structure of VSV-G in the V-LV and its desialylation by sialidase A. In V/HN-LV, SeV-HN cleaves sialic acid from VSV-G. (<b>F</b>) The LV particles obtained by ultracentrifugation of the viral supernatant were treated with sialidase A and subjected to western blotting using VSV-G, whole SeV, and HIV1 p24 antibodies. Untreated V-LV and V/HN-LV were included as negative and positive controls, respectively. (<b>G</b>,<b>H</b>) Summary graphs of the SeV-HN/p24 and VSV-G/p24 ratios. (<b>I</b>) Functional assay of the desialylated V-LV particles at 800 PP/cell. Scale bar, 100 µm. Data are expressed as the mean ± SD with biological replicates (n = 3). Ordinary two-way ANOVA with Tukey post-hoc test was conducted for (<b>B</b>,<b>C</b>) and ordinary one-way ANOVA with Tukey post-hoc test was conducted for (<b>G</b>–<b>I</b>). **** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.01, and * <span class="html-italic">p</span> &lt; 0.05; ns, not significant.</p>
Full article ">Figure 5
<p>N-glycolylneuraminic acid (Neu5Gc) inhibits V/HN-LV infection. (<b>A</b>) The CMAH enzyme converts CMP-Neu5Ac to CMP-Neu5Gc and is a pseudogene in humans and has thereby lost its activity. (<b>B</b>) HEK293FT cells were infected with LV particles at 400 PP/cell after feeding with 1.0 mM Neu5Ac or Neu5Gc for 9 days. The GFP-positive cells were evaluated by flow cytometry 2 days after transduction. (<b>C</b>) Neu5Gc expression was stained on WT-HEK293FT and CMAH-HEK293FT cells using an anti-Neu5Gc antibody. (<b>D</b>,<b>E</b>) The infection efficacy of LV particles on WT-HEK293FT and CMAH-HEK293FT cells was evaluated by (<b>D</b>) microscopic observation 48 h post-infection at 800 PP/cell and (<b>E</b>) flow cytometry analyses 48 h post-infection at the indicated PP/cell. (<b>F</b>) Median fluorescence intensity (MFI) was determined by flow cytometry. Scale bar, 100 µm. Data are expressed as the mean ± SD with biological replicates (n = 3) and ordinary two-way ANOVA with Tukey post-hoc test was conducted. **** <span class="html-italic">p</span> &lt; 0.0001 and *** <span class="html-italic">p</span> &lt; 0.001; ns, not significant.</p>
Full article ">Figure 6
<p>Improvement in the transduction efficiency of VSV-G and SeV-HN pseudotyped LVs in human hematopoietic stem and progenitor cells. (<b>A</b>,<b>B</b>) Representative flow cytometry plots of the GFP-positive rate in (<b>A</b>) with the human phenotypic hematopoietic stem cell (pHSC) population, defined as CD34<sup>+</sup>, CD38−, CD45RA−, and CD90<sup>+</sup>, and (<b>B</b>) with the phenotypic multipotent progenitor (pMPP) cell population, defined as CD34<sup>+</sup>, CD38−, CD45RA−, and CD90−. Human cord blood cells were infected with the four types of concentrated LV particles at 50,000 PP/cell, with production as shown in <a href="#app1-viruses-16-00827" class="html-app">Table S5</a>. The GFP-positive cells (<b>C</b>,<b>D</b>) and propidium iodide (PI)-negative cells (<b>E</b>,<b>F</b>) were evaluated by flow cytometry 2 days after transduction. The gating strategy is available in <a href="#app1-viruses-16-00827" class="html-app">Supplemental Figure S7</a>. Data are expressed as the mean ± SD with technical replicates (n = 5) and ordinary one-way ANOVA with Tukey post-hoc test was conducted. **** <span class="html-italic">p</span> &lt; 0.0001; ns, not significant.</p>
Full article ">
16 pages, 4054 KiB  
Article
N-Acetylneuraminic Acid Inhibits Melanogenesis via Induction of Autophagy
by Kei Yoshikawa and Kazuhisa Maeda
Cosmetics 2024, 11(3), 82; https://doi.org/10.3390/cosmetics11030082 - 21 May 2024
Viewed by 914
Abstract
N-acetylneuraminic acid (Neu5Ac) is the predominant form of sialic acid present in the glossy swiftlet (Collocalia esculenta). It is also the only form of sialic acid detected in the human body. In this study, we investigated the mechanism underlying melanogenesis [...] Read more.
N-acetylneuraminic acid (Neu5Ac) is the predominant form of sialic acid present in the glossy swiftlet (Collocalia esculenta). It is also the only form of sialic acid detected in the human body. In this study, we investigated the mechanism underlying melanogenesis inhibition by Neu5Ac. We discovered that a reduction in tyrosinase protein levels led to an inhibition of melanin production by Neu5Ac. Additionally, the mRNA and protein levels of ubiquitin-specific protease (USP5) and microtubule-associated protein 1 light chain 3 (LC3)-II increased, while those of p62 decreased, indicating enhanced autophagic activity. Lysosomal cathepsin L2 protein levels also increased, and immunostaining revealed colocalization of lysosomal membrane protein (LAMP)-1 and tyrosinase. Additionally, levels of chaperonin containing T-complex polypeptide (CCT), implicated in increased autophagic flux, were elevated. Altogether, these findings suggest that tyrosinase-containing coated vesicles are transported by Neu5Ac into the autophagic degradation pathway, suppressing mature melanosome generation. This process involves increased USP5 levels preventing recognition of polyubiquitin by proteasomes. Furthermore, elevated CCT3 protein levels may enhance autophagic flux, leading to the incorporation of tyrosinase-containing coated vesicles into autophagosomes. These autophagosomes then fuse with lysosomes for cathepsin L2–mediated degradation. Thus, our findings suggest that Neu5Ac reduces tyrosinase activity and inhibits melanosome maturation by promoting selective autophagic degradation of abnormal proteins by p62. Full article
(This article belongs to the Special Issue 10th Anniversary of Cosmetics—Recent Advances and Perspectives)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of <span class="html-italic">N</span>-acetylneuraminic acid (Neu5Ac).</p>
Full article ">Figure 2
<p>Effect of Neu5Ac on cell proliferation and tyrosinase and melanin levels in B16 melanoma cells. (<b>a</b>) Effect of Neu5Ac on cell number and tyrosinase activity. (<b>b</b>) Effect of Neu5Ac on tyrosinase mRNA and protein levels. Mean ± SD, <span class="html-italic">n</span> = 4 or <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. (<b>c</b>) Effect of Neu5Ac on melanin levels.</p>
Full article ">Figure 2 Cont.
<p>Effect of Neu5Ac on cell proliferation and tyrosinase and melanin levels in B16 melanoma cells. (<b>a</b>) Effect of Neu5Ac on cell number and tyrosinase activity. (<b>b</b>) Effect of Neu5Ac on tyrosinase mRNA and protein levels. Mean ± SD, <span class="html-italic">n</span> = 4 or <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. (<b>c</b>) Effect of Neu5Ac on melanin levels.</p>
Full article ">Figure 3
<p>Effect of Neu5Ac on tyrosinase and LAMP-1 localization, using fluorescence immunostaining (multiple staining). Nuclei were stained with DAPI (blue), tyrosinase was labeled with an anti-tyrosinase antibody, and Alexa Fluor 488 secondary antibody (green), and LAMP-1 was labeled with an anti-LAMP1 antibody and Alexa Fluor 568 secondary antibody (red). Tyrosinase and LAMP-1 colocalization areas are indicated in yellow. The scale bar is 100 μm.</p>
Full article ">Figure 4
<p>Effect of Neu5Ac on cathepsin L2 levels, using fluorescence immunostaining (multiple staining method). The nuclei were stained with DAPI (blue). Cathepsin L2 was labeled with an anti-cathepsin L2 antibody and Alexa Fluor 488 secondary antibody (green). The scale bar is 100 μm.</p>
Full article ">Figure 5
<p>(<b>a</b>) Effect of Neu5Ac on LC3. (<b>b</b>) LC3-II/β-actin ratio was calculated by measuring the brightness of LC3-II and β-actin bands with ImageJ. mean ± SD, <span class="html-italic">n</span> = 3. (<b>c</b>) Effect of Neu5Ac on p62. (<b>d</b>) The brightness of p62 and β-actin bands was measured with ImageJ. The p62/β-actin ratio was calculated and plotted. mean ± SD, <span class="html-italic">n</span> = 3, *: <span class="html-italic">p</span> &lt; 0.05 vs. control, **: <span class="html-italic">p</span> &lt; 0.01 vs. control.</p>
Full article ">Figure 5 Cont.
<p>(<b>a</b>) Effect of Neu5Ac on LC3. (<b>b</b>) LC3-II/β-actin ratio was calculated by measuring the brightness of LC3-II and β-actin bands with ImageJ. mean ± SD, <span class="html-italic">n</span> = 3. (<b>c</b>) Effect of Neu5Ac on p62. (<b>d</b>) The brightness of p62 and β-actin bands was measured with ImageJ. The p62/β-actin ratio was calculated and plotted. mean ± SD, <span class="html-italic">n</span> = 3, *: <span class="html-italic">p</span> &lt; 0.05 vs. control, **: <span class="html-italic">p</span> &lt; 0.01 vs. control.</p>
Full article ">Figure 6
<p>Effect of Neu5Ac on p62 localization, using fluorescence immunostaining. The nuclei were stained with DAPI (blue). P62 was labeled with an anti-p62/SQSTM1antibody and Alexa Fluor 568 secondary antibody (red). The scale bar was 50 μm.</p>
Full article ">Figure 6 Cont.
<p>Effect of Neu5Ac on p62 localization, using fluorescence immunostaining. The nuclei were stained with DAPI (blue). P62 was labeled with an anti-p62/SQSTM1antibody and Alexa Fluor 568 secondary antibody (red). The scale bar was 50 μm.</p>
Full article ">Figure 7
<p>Effect of Neu5Ac on mRNA (<b>a</b>) and protein levels (<b>b</b>–<b>d</b>) of the ubiquitin-proteasome system-related factors USP5 and UBE1Y. Mean ± SD, <span class="html-italic">n</span> = 3, *: <span class="html-italic">p</span> &lt; 0.05 vs. control, **: <span class="html-italic">p</span> &lt; 0.01 vs. control.</p>
Full article ">Figure 7 Cont.
<p>Effect of Neu5Ac on mRNA (<b>a</b>) and protein levels (<b>b</b>–<b>d</b>) of the ubiquitin-proteasome system-related factors USP5 and UBE1Y. Mean ± SD, <span class="html-italic">n</span> = 3, *: <span class="html-italic">p</span> &lt; 0.05 vs. control, **: <span class="html-italic">p</span> &lt; 0.01 vs. control.</p>
Full article ">Figure 8
<p>Effect of Neu5Ac on mRNA levels (<b>a</b>) of molecular chaperone CCT and protein levels of CCT3 (<b>b</b>,<b>c</b>). Mean ± SD, <span class="html-italic">n</span> = 3, *: <span class="html-italic">p</span> &lt; 0.05 vs. control, **: <span class="html-italic">p</span> &lt; 0.01 vs. control.</p>
Full article ">Figure 8 Cont.
<p>Effect of Neu5Ac on mRNA levels (<b>a</b>) of molecular chaperone CCT and protein levels of CCT3 (<b>b</b>,<b>c</b>). Mean ± SD, <span class="html-italic">n</span> = 3, *: <span class="html-italic">p</span> &lt; 0.05 vs. control, **: <span class="html-italic">p</span> &lt; 0.01 vs. control.</p>
Full article ">
Back to TopTop