[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (28)

Search Parameters:
Keywords = MRL/lpr mice

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 4663 KiB  
Article
Microplastics Exposure Aggravates Synovitis and Pyroptosis in SLE by Activating NF-κB and NRF2/KEAP1 Signaling
by Wenxiang Zeng, Shiqiao He, Ying Zhao, Minjian Jiang, Wenla Wang, Limeng Yang, Weibin Du and Wei Zhuang
Toxics 2024, 12(12), 840; https://doi.org/10.3390/toxics12120840 - 22 Nov 2024
Viewed by 537
Abstract
Microplastics (MPs) represent an emerging pollutant capable of entering the human body through the respiratory and digestive systems, thereby posing significant health risks. Systemic lupus erythematosus (SLE) is a complex autoimmune disease that affects multiple organ systems, often presenting with polyarticular joint manifestations. [...] Read more.
Microplastics (MPs) represent an emerging pollutant capable of entering the human body through the respiratory and digestive systems, thereby posing significant health risks. Systemic lupus erythematosus (SLE) is a complex autoimmune disease that affects multiple organ systems, often presenting with polyarticular joint manifestations. Despite its relevance, there is currently limited research on the impact of MPs on lupus arthritis. This study aims to investigate the effects of MPs on joint inflammation in SLE. MRL/lpr mice exhibit SLE similar to that of humans. We administered either 0.5 mg/kg or 5 mg/kg of MPs to 8-week-old female MRL/lpr mice via oral ingestion. Our findings indicate that exposure to MPs can lead to synovial damage, adversely affecting the morphology and function of the knee joint, along with increased oxidative stress, apoptosis, synovial fibrosis, and the secretion of inflammatory cytokines. Notably, MPs significantly enhanced synovial cell pyroptosis by upregulating the expression of NLRP3, CASPASE-1, GSDMD, IL-1β, and IL-18. Mechanistic analyses further demonstrated that MPs exposure activates the NF-κB and NRF2/KEAP1 signaling pathways. Overall, our in vivo findings suggest that MPs exposure promotes synovial cell pyroptosis through increased oxidative stress and NF-κB signaling, thereby disrupting the structure and function of synovial tissue. This research provides new insights into the synovial damage associated with MPs exposure. Full article
(This article belongs to the Section Reproductive and Developmental Toxicity)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The MP structure damages the synovial structure and aggravate synovitis in SLE mice: (<b>A</b>) flow chart of the experimental design; (<b>B</b>) HE staining of mouse knee synovium, demonstrating thickening of synovial layers (Black arrow) and increased vasculature (Blue arrow); (<b>C</b>) histological scoring of the synovium based on HE staining; (<b>D</b>) MASSON’s trichrome staining of mouse knee synovium, indicating increased fiber content (Black arrow); (<b>E</b>–<b>H</b>) immunohistochemical staining illustrating the expression of MMP-13 and MMP-19 in the synovium of the knee joint of mice, along with quantification of their expression levels. Red arrowheads denote positively stained cells. Data are presented as mean ± standard deviation (SD). ns indicates no statistical significance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BL/6J group), with <span class="html-italic">n</span> = 6 per group.</p>
Full article ">Figure 2
<p>MPs promote synovial cell apoptosis in SLE mice: (<b>A</b>,<b>C</b>) immunofluorescence staining demonstrating the expression of CASPASE-3 and BCL-2 in the synovium of the knee joints of mice, along with (<b>B</b>,<b>D</b>) quantification of expression; (<b>E</b>) TUNEL staining and (<b>F</b>) quantification of the rate of TUNEL-positive cells. DAPI stains the nuclei blue, and white arrowheads indicate positively stained cells. Data are presented as mean ± SD. ns indicates no statistical significance. ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BL/6J group), <span class="html-italic">n</span> = 6 per group.</p>
Full article ">Figure 3
<p>MPs compound synovial inflammation in SLE mice. (<b>A</b>–<b>H</b>) Immunofluorescence staining illustrating the expression of IL-1β, IL-18, IL-6, and TNF-α in the synovium of the knee joint of mice, along with quantification of expression. DAPI stains the nuclei blue, and white arrowheads indicate positively stained cells. Data are presented as mean ± SD. ns indicates no statistical significance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BJ/6 group), with <span class="html-italic">n</span> = 6 per group.</p>
Full article ">Figure 4
<p>MPs aggravate synovial pyroptosis in SLE mice. (<b>A</b>–<b>C</b>) Immunofluorescence staining showing the expression of NLRP3, CASPASE-1, and GSDMD in the synovium of the knee joint of mice, along with (<b>D</b>–<b>F</b>) quantification of expression. DAPI stains nuclei blue, and white arrowheads indicate positively stained cells. Data are expressed as mean ± SD. ns indicates no statistical significance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BL/6J group), with <span class="html-italic">n</span> = 6 per group.</p>
Full article ">Figure 5
<p>Effects of MPs exposure on oxidative stress and the NF-κB signaling pathway in SLE mice. (<b>A</b>–<b>C</b>) Immunofluorescence staining was conducted to assess the expression of NRF2, KEAP1, and HO-1 in the synovium of the knee joints of mice, while (<b>H</b>–<b>J</b>) illustrates the quantification of these expressions. (<b>D</b>–<b>G</b>) Immunofluorescence staining for P65, P-P65, IκBα, and p-IκBα in the knee joint synovium of mice is presented, with (<b>K</b>–<b>N</b>) showing the corresponding quantification. DAPI was used to stain nuclei in blue, and white arrowheads indicate positively stained cells. Data are expressed as mean ± SD. ns indicates no statistical significance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (vs. 0 mg/kg group); ## <span class="html-italic">p</span> &lt; 0.01 (vs. C57BL/6J group), with <span class="html-italic">n</span> = 6 per group.</p>
Full article ">
13 pages, 2873 KiB  
Article
Translating Lupus: Comparative Transcriptional Profiles of Preclinical Lupus Models and Their Relevance to Human Disease
by James T. Parker, Ching-Yun Chang, Kara Kersjes, Ixavier A. Higgins, Andrew C. Vendel and William Y. Chang
Biology 2024, 13(10), 778; https://doi.org/10.3390/biology13100778 - 28 Sep 2024
Viewed by 1023
Abstract
Systemic lupus erythematosus (SLE) is a chronic, systemic autoimmune disease which can present with mixed organ involvement. Kidney involvement in lupus nephritis (LN) is a severe complication and major cause of mortality in SLE patients, second only to cardiovascular disease. While mouse models [...] Read more.
Systemic lupus erythematosus (SLE) is a chronic, systemic autoimmune disease which can present with mixed organ involvement. Kidney involvement in lupus nephritis (LN) is a severe complication and major cause of mortality in SLE patients, second only to cardiovascular disease. While mouse models have helped uncover some molecular pathways involved in SLE/LN, we need a better understanding of the connection of these pathways and the immune cells involved in disease pathogenesis to develop new and effective therapies. Furthermore, models used for studying SLE/LN in mice have a heterogeneous immune response and may not always represent disease manifestations observed in patients. Identifying models that have shared pathways with human disease would allow for better translation for developing effective SLE/LN therapies. The molecular pathways of five different SLE/LN models (MRL/lpr, poly (I:C)-induced, interferon-α-induced, bm12 GvHD, and spontaneous NZB/W F1) were compared to characterize the immune response in mouse kidneys. These models demonstrated varied magnitudes in immune responses and proportions of innate vs. adaptive cell involvement. These findings were compared to human molecular pathways and cell types from public databases, including the Accelerating Medicine Partnership–Systemic Lupus Erythematosus Program (AMP-SLE), to help corelate mechanisms involved in mouse models to human disease. Full article
(This article belongs to the Special Issue Immune Response Regulation in Animals)
Show Figures

Figure 1

Figure 1
<p>Analysis of autoimmunity- and inflammation-associated mRNA expression in kidney tissue harvested from animal models of SLE/LN. (<b>A</b>) DEGs in each animal model reported as both total count and percentage of all genes measured. (<b>B</b>) Venn diagram of significant genes identified in the mIFNα, poly (I:C) and MRL/lpr animal models of SLE/LN. Significance was determined using one-way ANOVA model at significance cutoff of adjusted <span class="html-italic">p</span>-value of 0.1 and at least 1.5-fold change between diseased and naïve mice. (<b>C</b>) Differential expression of <span class="html-italic">Stat1</span> mRNA between diseased and naïve mice for each animal model. Asterisks indicate significant differences. Bars indicate mean +/− standard deviation.</p>
Full article ">Figure 2
<p>Pathway analysis of DEGs in animal models of SLE/LN. (<b>A</b>) Heatmap of modulated pathways (columns) for each animal model (rows). Colors binned by number of DEGs as a percentage of total genes for each pathway. (<b>B</b>) Heatmap of selected individual gene expression changes (columns) for genes in the Type I interferon signaling pathway, for each animal model (rows). Colors binned by fold changes (diseased vs. naïve mice) for each gene.</p>
Full article ">Figure 3
<p>Comparing gene signatures of SLE/LN human bulk kidney tissue or scRNA-seq with SLE/LN animal model gene expression profiles. (<b>A</b>) Heatmap of associations between human bulk tissue and SLE/LN animal models (rows) for selected pathways (columns). (<b>B</b>) Heatmap of associations between human scRNA-seq cell types and SLE/LN animal models (rows) for selected pathways (columns). (<b>C</b>) Association plots for human bulk tissue (left, <span class="html-italic">y</span>-axis) or human CD4 T cells from scRNA-seq (right, <span class="html-italic">y</span>-axis) with animal model tissue (<span class="html-italic">x</span>-axis) for Type I interferon signaling pathway genes. Proportional association scores indicated for each comparison (top right). (<b>D</b>) Association plots for human bulk tissue (left, <span class="html-italic">y</span>-axis) or human NK cells from scRNA-seq (right, <span class="html-italic">y</span>-axis) with animal model tissue (<span class="html-italic">x</span>-axis) for cytotoxicity pathway genes. Proportional association scores indicated for each comparison (top right).</p>
Full article ">
20 pages, 3630 KiB  
Article
Tissue Kallikrein-1 Suppresses Type I Interferon Responses and Reduces Depressive-Like Behavior in the MRL/lpr Lupus-Prone Mouse Model
by Priyanka S. Bhoj, Cassandra Nocito, Namdev S. Togre, Malika Winfield, Cody Lubinsky, Sabeeya Khan, Nikhita Mogadala, Alecia Seliga, Ellen M. Unterwald, Yuri Persidsky and Uma Sriram
Int. J. Mol. Sci. 2024, 25(18), 10080; https://doi.org/10.3390/ijms251810080 - 19 Sep 2024
Cited by 1 | Viewed by 1258
Abstract
Excessive production and response to Type I interferons (IFNs) is a hallmark of systemic lupus erythematosus (SLE). Neuropsychiatric lupus (NPSLE) is a common manifestation of human SLE, with major depression as the most common presentation. Clinical studies have demonstrated that IFNα can cause [...] Read more.
Excessive production and response to Type I interferons (IFNs) is a hallmark of systemic lupus erythematosus (SLE). Neuropsychiatric lupus (NPSLE) is a common manifestation of human SLE, with major depression as the most common presentation. Clinical studies have demonstrated that IFNα can cause depressive symptoms. We have shown that the kallikrein–kinin system (KKS) [comprised of kallikreins (Klks) and bradykinins] and angiotensin-converting enzyme inhibitors suppressed Type I IFN responses in dendritic cells from lupus-prone mice and human peripheral blood mononuclear cells. Tissue Klk genes are decreased in patients with lupus, and giving exogenous Klk1 ameliorated kidney pathology in mice. We retro-orbitally administered mouse klk1 gene-carrying adenovirus in the Murphy Roths Large lymphoproliferative (MRL/lpr) lupus-prone mice at early disease onset and analyzed immune responses and depressive-like behavior. Klk1 improved depressive-like behavior, suppressed interferon-responsive genes and neuroinflammation, and reduced plasma IFNα levels and proinflammatory cytokines. Klk1 also reduced IFNAR1 and JAK1 protein expression, important upstream molecules in Type I IFN signaling. Klk1 reduced bradykinin B1 receptor expression, which is known to induce proinflammatory response. Together, these findings suggest that Klk1 may be a potential therapeutic candidate to control IFNα production/responses and other inflammatory responses in SLE and NPSLE. Full article
(This article belongs to the Topic Inflammation: The Cause of all Diseases 2.0)
Show Figures

Figure 1

Figure 1
<p>Administration of <span class="html-italic">Ad-klk1</span> increases Klk1 protein expression (<b>A</b>) Western blot of Klk1 protein (MW: 28 kDa) and (<b>B</b>) immunohistochemical results (indicated by pink staining) of Klk1 protein expression in the kidney of the spontaneous cohort. Images were taken at 40× magnification and analyzed using ImageJ (1.54f software). The results are expressed as mean ±SEM. An unpaired <span class="html-italic">t</span>-test was used; * <span class="html-italic">p</span> ≤ 0.05 and *** <span class="html-italic">p</span> ≤ 0.001 compared to the Ad-GFP group; <span class="html-italic">n =</span> 7–8 mice/group.</p>
Full article ">Figure 2
<p>Tissue Klk1 decreases IRGs and <span class="html-italic">hmgb1</span> gene expression in the brain and periphery: (<b>A</b>) Spontaneous MRL/lpr mice and (<b>B</b>) IFNα-induced MRL/lpr mice. The data, obtained by qPCR, were analyzed using an unpaired <span class="html-italic">t</span>-test and are expressed as the mean ±SEM of the fold change in gene expression in the Ad-Klk1 group compared to the Ad-GFP group (marked as a solid black line at 1.0 on the Y axis). * <span class="html-italic">p</span> ≤ 0.05 and ** <span class="html-italic">p</span> ≤ 0.01 compared to the Ad-GFP group; <span class="html-italic">n =</span> 7–8 mice/group in the spontaneous cohort, and <span class="html-italic">n =</span> 5 mice/group in the IFNα-induced cohort.</p>
Full article ">Figure 3
<p>Tissue Klk1 reduces inflammatory cytokine levels in the plasma: (<b>A</b>) Spontaneous MRL/lpr mice and (<b>B</b>) IFNα-induced MRL/lpr mice. Multiplex cytokine analysis was performed using the MSD ELISA kit. The data were analyzed using an unpaired <span class="html-italic">t</span>-test and are expressed as the mean ±SEM. * <span class="html-italic">p</span> ≤ 0.05 compared to the Ad-GFP group; <span class="html-italic">n =</span> 7–8 mice/group in the spontaneous cohort, and <span class="html-italic">n =</span> 5 mice/group in the IFNα-induced cohort.</p>
Full article ">Figure 4
<p>Tissue Klk1 decreases plasma IFNα levels and proteins in Type I IFN signaling: (<b>A</b>) Plasma IFNα levels in the spontaneous MRL/lpr mice were measured using the high sensitivity Verikine ELISA kit and are expressed as the mean ±SEM of IFNα levels (pg/mL). Western blot analysis of IFNAR1 (MW: 64 kDa) and JAK1 (MW: 80 kDa) protein levels in the (<b>B</b>) kidney and (<b>C</b>) spleen of spontaneous MRL/lpr mice. Data were analyzed using an unpaired <span class="html-italic">t</span>-test and are expressed as the mean ±SEM of the fold change in protein levels in the Ad-Klk1 group compared to the Ad-GFP group (marked as a solid black line at 1.0 on the Y axis). * <span class="html-italic">p</span> ≤ 0.05 and ** <span class="html-italic">p</span> ≤ 0.01 compared to the Ad-GFP group; <span class="html-italic">n =</span> 7–8 mice/group.</p>
Full article ">Figure 5
<p>Tissue Klk1 decreases depressive-like behavior and depression-related gene expression in the spontaneous MRL/lpr mice: (<b>A</b>) Tail suspension test (TST). (<b>B</b>) Plasma serotonin levels were estimated by using the ELISA kit. (<b>C</b>) Gene expression analysis of serotonin regulators in the brain; the results are expressed as the mean ±SEM of the fold change in gene expression in the Ad-Klk1 group compared to the Ad-GFP group (marked as a solid black line at 1.0 on the Y axis). (<b>D</b>) Correlation of the immobility time in the TST with plasma IFNα levels and serotonin-related markers (brain <span class="html-italic">isg15</span>, <span class="html-italic">5htt</span>, and <span class="html-italic">tph-2</span>). The data were analyzed using an unpaired <span class="html-italic">t</span>-test and * <span class="html-italic">p</span> ≤ 0.05 compared to the Ad-GFP group; <span class="html-italic">n =</span> 7–8 mice/group.</p>
Full article ">Figure 6
<p>Tissue Klk1 reduced microglial inflammatory state and number. Immunohistochemical staining of IBA-1 in microglia. Data were analyzed using an unpaired <span class="html-italic">t</span>-test and are expressed as the mean ±SEM of the percentage area of IBA-1 staining in the cortex region of the brain at 40× magnification (brown staining, indicated by yellow arrows) using ImageJ (1.54f software). * <span class="html-italic">p</span> ≤ 0.05 compared to Ad-GFP group; <span class="html-italic">n =</span> 7–8 mice/group.</p>
Full article ">Figure 7
<p>Tissue Klk1 decreases C3 and IgG protein levels in the kidney: Western blot analysis of C3 (MW: 50 kDa) and IgG (MW: 50 kDa) protein levels in the kidney of spontaneous MRL/lpr mice. Data were analyzed using an unpaired <span class="html-italic">t</span>-test and are expressed as the mean ±SEM of the fold change in protein levels in the Ad-Klk1 group compared to the Ad-GFP group (marked as a solid black line at 1.0 on the Y axis). * <span class="html-italic">p</span> ≤ 0.05 compared to the Ad-GFP group; <span class="html-italic">n =</span> 7–8 mice/group.</p>
Full article ">Figure 8
<p>Tissue Klk1 alters BK receptor expression in the spontaneous MRL/lpr mice: (<b>A</b>) Fold change in <span class="html-italic">b1r</span> and <span class="html-italic">b2r</span> gene expression in the brain of the Ad-Klk1 group compared to those of the Ad-GFP group (marked as a solid black line at 1.0 on the Y axis), as measured by qPCR. Fold change in B1R (MW: 40 kDa) and B2R (MW: 50 kDa) protein levels in the (<b>B</b>) kidneys and (<b>C</b>) spleen of the Ad-Klk1 group compared to those of the Ad-GFP group (marked as a solid black line at 1.0 on the Y axis), as measured by western blot. The results are expressed as mean ±SEM. An unpaired <span class="html-italic">t</span>-test was used; * <span class="html-italic">p</span> ≤ 0.05 and <span class="html-italic">p</span> ≤ 0.001 compared to the Ad-GFP group; <span class="html-italic">n =</span> 7–8 mice/group.</p>
Full article ">Figure 9
<p>Scheme of the experiment: One cohort of nine-week-old MRL/lpr mice was administered with IFNα- or saline-carrying osmotic pump (IFNα-induced response) on day −2, followed by the retro-orbital administration of <span class="html-italic">Ad-klk1</span> or <span class="html-italic">Ad-gfp</span> vector on day 0. The spontaneous cohort received <span class="html-italic">Ad-klk1</span> or control (<span class="html-italic">Ad-gfp</span>) vector only. Behavioral studies were performed on day 15, and the mice were sacrificed on day 17 to collect the brain, spleen, kidneys, and blood.</p>
Full article ">Figure 10
<p>Tissue Klk1 suppresses Type I IFN responses through IFNAR1, BRs, and PAR2 and reduces depressive-like behavior in the MRL/lpr lupus-prone mice: The present study demonstrates that tissue Klk1 administration reduces IFNα, IFNAR1, JAK1, and IRF7, consequently suppressing IRG expression. Our previous study [<a href="#B11-ijms-25-10080" class="html-bibr">11</a>] has shown a reduction in signal transducer and activator of transcription (STAT) phosphorylation (shown in blue color) following bradykinin (BK) administration, which, along with other pathway proteins (shown in grey color), will be studied in detail in the future. We observed a consequential reduction in the levels of tryptophan hydroxylase 2 (TPH2, a rate-limiting enzyme in serotonin biosynthesis), serotonin (5-HT), serotonin transporter (5-HTT), and 5-hydroxytryptamine receptor 2A (HTR2A, a serotonin receptor). We also observed depressive-like behaviors in MRL/lpr mice. Alleviation of inflammatory and IFN responses may be mediated via the alteration in BK receptors (B1R and B2R) and protease-activated receptor-2 (PAR2) expression; the mechanisms need to be studied in detail (indicated with dashed arrows). The therapeutic effects of Klk1 are shown in green notation, and SLE and NPSLE pathogenesis are represented in red notation.</p>
Full article ">
17 pages, 1589 KiB  
Article
Effect of Spermidine on Endothelial Function in Systemic Lupus Erythematosus Mice
by Hyoseon Kim and Michael P. Massett
Int. J. Mol. Sci. 2024, 25(18), 9920; https://doi.org/10.3390/ijms25189920 - 14 Sep 2024
Viewed by 1213
Abstract
Endothelial dysfunction is common in Systemic Lupus Erythematosus (SLE), even in the absence of cardiovascular disease. Evidence suggests that impaired mitophagy contributes to SLE. Mitochondrial dysfunction is also associated with impaired endothelial function. Spermidine, a natural polyamine, stimulates mitophagy by the PINK1–parkin pathway [...] Read more.
Endothelial dysfunction is common in Systemic Lupus Erythematosus (SLE), even in the absence of cardiovascular disease. Evidence suggests that impaired mitophagy contributes to SLE. Mitochondrial dysfunction is also associated with impaired endothelial function. Spermidine, a natural polyamine, stimulates mitophagy by the PINK1–parkin pathway and counters age-associated endothelial dysfunction. However, the effect of spermidine on mitophagy and vascular function in SLE has not been explored. To address this gap, 9-week-old female lupus-prone (MRL/lpr) and healthy control (MRL/MpJ) mice were randomly assigned to spermidine treatment (lpr_Spermidine and MpJ_Spermidine) for 8 weeks or as control (lpr_Control and MpJ_Control). lpr_Control mice exhibited impaired endothelial function (e.g., decreased relaxation to acetylcholine), increased markers of inflammation, and lower protein content of parkin, a mitophagy marker, in the thoracic aorta. Spermidine treatment prevented endothelial dysfunction in MRL-lpr mice. Furthermore, aortas from lpr_Spermidine mice had lower levels of inflammatory markers and higher levels of parkin. Lupus phenotypes were not affected by spermidine. Collectively, these results demonstrate the beneficial effects of spermidine treatment on endothelial function, inflammation, and mitophagy in SLE mice. These results support future studies of the beneficial effects of spermidine on endothelial dysfunction and cardiovascular disease risk in SLE. Full article
Show Figures

Figure 1

Figure 1
<p>Spermidine prevents endothelial dysfunction in lupus mice. Relaxation responses to increasing concentrations of ACh (<b>A</b>) and SNP (<b>B</b>) in thoracic aortas from MRL/lpr and MRL/MpJ mice with and without spermidine treatment. Values are mean ± SEM, n = 11–15 per group. BL, baseline after 70% maximal contraction with phenylephrine. * significant difference between lpr_Control and MpJ_Control, <span class="html-italic">p</span> &lt; 0.05. # significant difference between lpr_Control and lpr_Spermidine, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Effect of spermidine on endothelial nitric oxide synthase (eNOS). Densitometry analysis of the Western Blot and mRNA expression for p-eNOS/eNOS (<b>A</b>), total eNOS (<b>B</b>), and NOS3 (<b>C</b>) in thoracic and abdominal aortas from MRL/lpr and MRL/MpJ mice with and without spermidine treatment. (<b>D</b>) Western Blot images for phosphorylated eNOS, total eNOS, and total protein by Ponceau stain in thoracic aorta. Values are mean ± SEM. n = 3–5 mice per group.</p>
Full article ">Figure 3
<p>Negative correlation between anti-dsDNA Ab levels and maximal responses to ACh (%). The box contains the correlation coefficient (r) and <span class="html-italic">p</span> value.</p>
Full article ">Figure 4
<p>SLE mice have impaired mitophagy in the aorta and liver. Densitometry analysis of the Western Blot for parkin (<b>A</b>,<b>B</b>) and LC3II/I (<b>C</b>,<b>D</b>) in thoracic aortas (<b>left</b>: <b>A</b>,<b>C</b>) and livers (<b>right</b>: <b>B</b>,<b>D</b>) from MRL/lpr and MRL/MpJ mice with and without spermidine treatment. (<b>E</b>) Western Blot images of parkin, LC3II, LC3I, and total protein by Ponceau stain. Values are mean ± SEM, n = 3–6 mice per group. * significantly different from MpJ_Control, <span class="html-italic">p</span> &lt; 0.05. # significantly different from lpr_Control, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Vascular inflammatory markers are elevated in lupus mice. Densitometry analysis of the Western Blot (<b>A</b>) and mRNA expression (<b>B</b>) for Vcam1 in thoracic and abdominal aortas from MRL/lpr and MRL/MpJ mice with and without spermidine treatment. (<b>C</b>) Western Blot images of Vcam1 and total protein by Ponceau stain. Values are mean ± SEM. n = 4–7 mice per group. * significantly different from MpJ_Control, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Interferon regulatory factor 1 (<span class="html-italic">Irf-1</span>) gene expression in lupus mice. mRNA expression of interferon regulatory factor 1 (<span class="html-italic">Irf1</span>) shown in the spleen (<b>A</b>) and liver (<b>B</b>) from MRL/lpr and MRL/MpJ mice with and without spermidine treatment. Values are mean ± SEM. n = 3–4 mice per group. * significantly different from MpJ_Control, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
22 pages, 943 KiB  
Review
Emerging Molecular and Synaptic Targets for the Management of Chronic Pain Caused by Systemic Lupus Erythematosus
by Han-Rong Weng
Int. J. Mol. Sci. 2024, 25(7), 3602; https://doi.org/10.3390/ijms25073602 - 22 Mar 2024
Cited by 1 | Viewed by 2023
Abstract
Patients with systemic lupus erythematosus (SLE) frequently experience chronic pain due to the limited effectiveness and safety profiles of current analgesics. Understanding the molecular and synaptic mechanisms underlying abnormal neuronal activation along the pain signaling pathway is essential for developing new analgesics to [...] Read more.
Patients with systemic lupus erythematosus (SLE) frequently experience chronic pain due to the limited effectiveness and safety profiles of current analgesics. Understanding the molecular and synaptic mechanisms underlying abnormal neuronal activation along the pain signaling pathway is essential for developing new analgesics to address SLE-induced chronic pain. Recent studies, including those conducted by our team and others using the SLE animal model (MRL/lpr lupus-prone mice), have unveiled heightened excitability in nociceptive primary sensory neurons within the dorsal root ganglia and increased glutamatergic synaptic activity in spinal dorsal horn neurons, contributing to the development of chronic pain in mice with SLE. Nociceptive primary sensory neurons in lupus animals exhibit elevated resting membrane potentials, and reduced thresholds and rheobases of action potentials. These changes coincide with the elevated production of TNFα and IL-1β, as well as increased ERK activity in the dorsal root ganglion, coupled with decreased AMPK activity in the same region. Dysregulated AMPK activity is linked to heightened excitability in nociceptive sensory neurons in lupus animals. Additionally, the increased glutamatergic synaptic activity in the spinal dorsal horn in lupus mice with chronic pain is characterized by enhanced presynaptic glutamate release and postsynaptic AMPA receptor activation, alongside the reduced activity of glial glutamate transporters. These alterations are caused by the elevated activities of IL-1β, IL-18, CSF-1, and thrombin, and reduced AMPK activities in the dorsal horn. Furthermore, the pharmacological activation of spinal GPR109A receptors in microglia in lupus mice suppresses chronic pain by inhibiting p38 MAPK activity and the production of both IL-1β and IL-18, as well as reducing glutamatergic synaptic activity in the spinal dorsal horn. These findings collectively unveil crucial signaling molecular and synaptic targets for modulating abnormal neuronal activation in both the periphery and spinal dorsal horn, offering insights into the development of analgesics for managing SLE-induced chronic pain. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration for altered membrane properties in nociceptive primary sensory neurons in the DRG in lupus mice with chronic pain. Lupus mice with chronic pain exhibit elevated resting membrane potentials, a reduced action potential (AP) activating thresholds and rheobases, and lowered capacitance, while displaying no changes in AP half-duration and amplitude compared to normal control mice. These changes result in hyperexcitability in nociceptive primary sensory neurons in lupus mice [<a href="#B28-ijms-25-03602" class="html-bibr">28</a>].</p>
Full article ">Figure 2
<p>A schematic illustration of the primary signaling molecular and synaptic mechanisms governing the excitability of DRG nociceptive sensory neurons and the glutamatergic synaptic activity in the spinal dorsal horn of mice with chronic pain induced by SLE. In lupus mice with chronic pain, DRG nociceptive neurons exhibit increased excitability. This is attributed to the elevated production of TNFα and IL-1β in the DRG, which suppress AMPK activity. Consequently, AMPK suppression leads to inhibition of the Na<sup>+</sup>-K<sup>+</sup> ATPase pump in the plasma membrane, raising the resting membrane potential. Additionally, reduced AMPK activity activates ERK, phosphorylating Nav. 1.7 channels and lowering their activation threshold. The combination of elevated resting membrane potential and reduced Nav. 1.7 activation threshold decreases neuronal rheobase, resulting in hyperexcitability of DRG nociceptive sensory neurons (i.e., peripheral sensitization). Furthermore, in the spinal dorsal horn of lupus mice with chronic pain, there is increased glutamatergic synaptic activity. The elevated expression of IL-1β, IL-18, CSF-1, and thrombin, along with reduced AMPK activity, contributes to this phenomenon. The increased activation of CSF-1 receptors (CSF-1-R) on microglia promotes IL-1β and IL-18 production. Presynaptic glutamate release is augmented by IL-1β, IL-18, CSF-1, thrombin, and reduced AMPK activity. IL-1β enhances presynaptic glutamate release by activating presynaptic NMDA receptors (NMDA-R). IL-1β also potentiates AMPA receptor (AMPA-R) and NMDA receptor (NMDA -R) activity in postsynaptic neurons. Moreover, the activation of IL-1β receptors (IL-1β-R) and thrombin receptor (PAR1) on astrocytes suppresses glial glutamate transporter (GLT-1) function by inhibiting AMPK activity. The activation of PAR1 or reduced AMPK activity at postsynaptic neurons leads to enhanced AMPA receptor activity in postsynaptic neurons. Overall, the increased glutamatergic activity in the spinal dorsal horn indicates central sensitization in lupus mice with chronic pain. Finally, the activation of GPR109A by the selective GPR109A agonist (MK1903) on microglia suppresses the enhanced glutamatergic activity by suppressing the production of IL-1β and IL-18. Symbols: ↑ increased; ↓ decreased.</p>
Full article ">
19 pages, 16779 KiB  
Article
Reduced Renal CSE/CBS/H2S Contributes to the Progress of Lupus Nephritis
by Xuan Wang, Tao Lin, Yifei He, Yueyuan Zhou, Yi Peng, Weiru Zhang and Xin Ni
Biology 2023, 12(2), 318; https://doi.org/10.3390/biology12020318 - 16 Feb 2023
Cited by 1 | Viewed by 2412
Abstract
The molecular mechanisms underlying lupus nephritis (LN) pathogenesis are not fully understood. Hydrogen sulfide (H2S) is involved in many pathological and physiological processes. We sought to investigate the roles of H2S in LN pathogenesis. H2S synthase cystathionine–lyase (CSE) and cystathionine–synthetase (CBS) expression was [...] Read more.
The molecular mechanisms underlying lupus nephritis (LN) pathogenesis are not fully understood. Hydrogen sulfide (H2S) is involved in many pathological and physiological processes. We sought to investigate the roles of H2S in LN pathogenesis. H2S synthase cystathionine–lyase (CSE) and cystathionine–synthetase (CBS) expression was downregulated in renal tissues of patients with LN and their levels were associated with LN’s prognosis using the Nephroseq database. Reduced CSE and CBS protein expression in kidney tissues of LN patients and MRL/lpr mice were confirmed by immunohistochemistry. CSE and CBS mRNA levels were reduced in MRL/lpr and pristine- and R848-induced lupus mice. Given that H2S exerts an anti-inflammatory role partly via regulating inflammatory transcription factors (TFs), we analyzed hub TFs by using a bioinformatics approach. It showed that STAT1, RELA, and T-cell-related signaling pathways were enriched in LN. Increased STAT1 and RELA expression were confirmed in renal tissues of LN patients. Treatment of MRL/lpr and pristine mice with H2S donors alleviated systemic lupus erythematosus (SLE) phenotypes and renal injury. H2S donors inhibited RELA level and T-cell infiltration in the kidneys of MRL/lpr and pristine mice. Our data indicated that CSE/CBS/H2S contributes to LN pathogenesis. Supplementation of H2S would be a potential therapeutic strategy for LN. Full article
(This article belongs to the Special Issue Autoimmune Diseases: Molecular and Cellular Mechanisms)
Show Figures

Figure 1

Figure 1
<p>Downregulation of hydrogen sulfide synthetase in the kidneys of lupus nephritis patients was associated with poor renal prognosis. (<b>A</b>,<b>B</b>) Gene expression of renal CSE in Glom and TubInt of healthy living donors and LN patients (Nephroseq database). (<b>C</b>,<b>D</b>) Gene expression of renal CBS in Glom and TubInt of healthy living donors and LN patients. (<b>E</b>,<b>F</b>) Correlation of renal TubInt CSE gene expression with serum creatinine and GFR in patients with LN in Nephroseq database. (<b>G</b>,<b>H</b>) Correlation of renal Glom CBS gene expression with serum creatinine and GFR in patients with LN in Nephroseq database.</p>
Full article ">Figure 2
<p>CSE and CBS were decreased in the kidney tissue of lupus mice. (<b>A</b>) Representative immunohistochemical staining images of CSE and CBS from human renal-puncture tissue (400×). Glom: glomeruli; TubInt: tubulointerstitium; <span class="html-italic">n</span>: total number of data; Ctrl: normal or minimal change kidney of the donor. (<b>B</b>) Representative immunofluorescence staining images of CSE, CBS, DAPI, and αSMA (a marker of glomerular mesangial cells and renal interstitial vascular smooth muscle cells) from the kidney of MRL/mpj and MRL/lpr mice (200×); the white arrows and enlarge box in the upper right corner of pictures refer to the glomerulus. (<b>C</b>) Representative immunohistochemical staining images of CSE and CBS from the kidney of MRL/mpj and MRL/lpr mice (40× and 400×). (<b>D</b>) mRNA expression of CSE in 16-week-old MRL/mpj and MRL/lpr mice. (<b>E</b>) mRNA expression of CSE in control Balb/c and pristane-induced Balb/c mice. (<b>F</b>) mRNA expression of CSE in control Balb/c and R848-induced Balb/c mice. (<b>G</b>) mRNA expression of CBS in 16-week-old MRL/mpj and MRL/lpr mice. (<b>H</b>) mRNA expression of CBS in control Balb/c and pristane-induced Balb/c mice. (<b>I</b>) mRNA expression of CBS in control Balb/c and R848-induced Balb/c mice. (<b>J</b>,<b>K</b>) mRNA expression of CSE and CBS in 8-week-old (pre-diseased) and 16-week-old (severely diseased) MRL/lpr mice. *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Screen of key TFs in renal tissue of patients with LN and its correlation with CBS and CSE. The Venn diagram of the differential TFs in the tubulointerstitium (<b>A</b>) and glomeruli (<b>B</b>), based on GSE32591, GSE113342, and human TFDB. (<b>C</b>) Red represents upregulation and blue represents downregulation for the differentially expressed TF genes (DETFGs) in the tubulointerstitium and glomeruli. (<b>D</b>) The interaction of hub transcription factors in the tubulointerstitium: the redder the color, the higher the degree through modules cytohHubba for sorting top hub genes. (<b>E</b>) The interaction of hub transcription factors in the glomeruli. (<b>F</b>–<b>H</b>) Correlation analysis between gene expression of CSE, CBS, and DETFGs. TUB/TubInt, tubulointerstitium; GLOM, glomeruli; *, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>The expression of p65 and STAT1 in kidney tissue of LN patients. (<b>A</b>,<b>B</b>) Representative images of p65, p-p65, STAT1, and p-STAT1 staining in renal tissues of control and patients with LN (400×). TubInt, tubulointerstitium; Glom, glomeruli.</p>
Full article ">Figure 5
<p>KEGG enrichment analysis of DETFGs in the tubulointerstitial and glomeruli of renal tissue of LN. (<b>A</b>) KEGG enrichment analysis of DETFGs in the tubulointerstitium; red and blue markers represent the signaling pathways associated with T cells and B cells, respectively. (<b>B</b>) KEGG enrichment analysis of DETFGs in glomeruli.</p>
Full article ">Figure 6
<p>Immune cell subtype infiltration in the tubulointerstitial and glomeruli of renal tissue of LN. (<b>A</b>,<b>B</b>) The proportion of the 26 types of immune cell infiltration in the tubulointerstitium (<b>A</b>) and glomeruli (<b>B</b>). (<b>C</b>,<b>D</b>) Association between TFs and immune cell populations in LN. The association of TFs with immune cell populations in the tubulointerstitium (<b>C</b>) and glomeruli (<b>D</b>). Tgd, gamma delta T cells; Tr1, type 1 regulatory T cells; nTreg, natural T-regulatory cells; Tcm, central memory T; *, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>NaHS-attenuated pristane-induced renal injury, splenic proliferation, and arthritis. (<b>A</b>) Representative images of mouse renal tissues after HE staining (400×). (<b>B</b>) The ratio of urine protein and urine creatinine from mice was measured. Data are represented as the means ± SEM (<span class="html-italic">n</span> = 5). * <span class="html-italic">p</span> &lt; 0.01, vs. control group. # <span class="html-italic">p</span> &lt; 0.01, vs. pristane-induced lupus group. (<b>C</b>,<b>D</b>) Images of spleens and the lower ankle from control, pristane-induced with or without NaHS mice are shown. (<b>E</b>) Representative scan images of mouse lower ankle after HE staining.</p>
Full article ">Figure 8
<p>The H2S donors, GYY4137 and NaHS, partially attenuated renal injury in MRL/lpr mice. (<b>A</b>) Representative images of mouse renal tissues after HE staining in GYY4137-treated mice (400×). (<b>B</b>–<b>E</b>) The serum levels of creatinine, urea nitrogen, uric acid, and albumin were detected upon GYY4137 injection. (<b>F</b>,<b>G</b>) The ratio of spleen weight and body weight and representative images of the spleen have been shown. Data are represented as the means ± SEM (<span class="html-italic">n</span> = 6). * <span class="html-italic">p</span> &lt; 0.05. (<b>H</b>) Representative images of mouse renal tissues after HE staining in NaHS-treated mice. (<b>I</b>) The ratio of urine protein and urine creatinine from mice was measured. (<b>J</b>,<b>K</b>) The ratio of spleen or cervical lymph node weight and body weight was calculated.</p>
Full article ">Figure 9
<p>H2S donor recovers the reduced expression of CSE and CBS in MRL/lpr mice. (<b>A</b>) Representative images of mouse renal tissues after IHC staining of CSE and CBS in MRL/lpr and MRL/mpj mice (400×). (<b>B</b>,<b>C</b>) Representative images of mouse renal tissues after IF staining of CSE, CBS, and DAPI in MRL/lpr and MRL/mpj mice (200×); the white arrow refers to the glomerulus.</p>
Full article ">Figure 10
<p>H2S supplementation reduces the expression of p65 and T-cell infiltration in the kidney of lupus mice. (<b>A</b>,<b>B</b>) Representative images of mouse renal tissues after IHC staining of p65 and p-p65 in pristane-induced mice (400×). (<b>C</b>) A model depicting the potential role of the H2S donor in the attenuation of LN.</p>
Full article ">
13 pages, 3042 KiB  
Article
Intraperitoneally Delivered Umbilical Cord Lining Mesenchymal Stromal Cells Improve Survival and Kidney Function in Murine Lupus via Myeloid Pathway Targeting
by Alvin Wen Choong Chua, Dianyang Guo, Jia Chi Tan, Frances Ting Wei Lim, Chee Tian Ong, Jeyakumar Masilamani, Tony Kiat Hon Lim, William Ying Khee Hwang, Ivor Jiun Lim, Jinmiao Chen, Toan Thang Phan and Xiubo Fan
Int. J. Mol. Sci. 2023, 24(1), 365; https://doi.org/10.3390/ijms24010365 - 26 Dec 2022
Cited by 2 | Viewed by 2523
Abstract
To determine the therapeutic efficacy of human umbilical cord lining mesenchymal stromal cells (CL-MSCs) (US Patent number 9,737,568) in lupus-prone MRL/lpr (Faslpr) mice and elucidate its working mechanisms. A total of 4 doses of (20–25) × 106 cells/kg of CL-MSCs [...] Read more.
To determine the therapeutic efficacy of human umbilical cord lining mesenchymal stromal cells (CL-MSCs) (US Patent number 9,737,568) in lupus-prone MRL/lpr (Faslpr) mice and elucidate its working mechanisms. A total of 4 doses of (20–25) × 106 cells/kg of CL-MSCs was given to 16-week-old female Faslpr mice by intraperitoneal injection. Three subsequent doses were given on 17 weeks, 18 weeks, and 22 weeks, respectively. Six-week-old Faslpr mice were used as disease pre-onset controls. Mice were monitored for 10 weeks. Mouse kidney function was evaluated by examining complement component 3 (C3) deposition, urinary albumin-to-creatinine ratio (ACR), and lupus nephritis (LN) activity and chronicity. Working mechanisms were elucidated by flow cytometry, Luminex/ELISA (detection of anti-dsDNA and isotype antibodies), and RNA sequencing. CL-MSCs improved mice survival and kidney function by reducing LN activity and chronicity and lymphocyte infiltration over 10 weeks. CL-MSCs also reduced urinary ACR, renal complement C3 deposition, anti-dsDNA, and isotype antibodies that include IgA, IgG1, IgG2a, IgG2b, and IgM. Immune and cytokine profiling demonstrated that CL-MSCs dampened inflammation by suppressing splenic neutrophils and monocytes/macrophages, reducing plasma IL-6, IL-12, and CXCL1 and stabilizing plasma interferon-γ and TNF-α. RNA sequencing further showed that CL-MSCs mediated immunomodulation via concerted action of pro-proinflammatory cytokine-induced chemokines and production of nitric oxide in macrophages. CL-MSCs may provide a novel myeloid (neutrophils and monocytes/macrophages)-targeting therapy for SLE. Full article
Show Figures

Figure 1

Figure 1
<p>Intraperitoneal CL-MSCs were able to improve mice survival and reduce disease activity: (<b>A</b>) Survival curves of Fas<sup>lpr</sup> mice treated with single and multiple doses of CL-MSCs. Twenty-four, 15, 7, and 10 mice were used in dPBS control, CL-MSCs (a total of 4 doses on 16-week-old, 17-week-old, 18-week-old, and 22-week-old mice), and CL-MSCs (single dose on 16-week-old mice) and CL-MSCs (a total of 10 doses weekly) groups. (<b>B</b>) Histopathological evaluation of LN activity and chronicity at 10 weeks PT. Three mice per group. (<b>C</b>) H&amp;E staining. Kidney sections from each group at 10-week PT. Scale bar: 100 μm. Results were expressed as mean ± SD. For multiple comparison, the significance was defined as * <span class="html-italic">p</span> &lt; 0.025; ** <span class="html-italic">p</span> &lt; 0.005 when n = 2. One-way ANOVA Dunnett’s test was used for multiple comparison with 95% confidence interval.</p>
Full article ">Figure 2
<p>Kidney function was improved by CL-MSCs treatment: (<b>A</b>) Urinary ACR changes at three weeks PT. Twelve, 7, and 9 mice were used for 6-week-old pre-onset mice, dPBS control, and CL-MSCs. (<b>B</b>) The number of glomerulus with complement C3 deposition in the kidneys at three weeks PT (40-fold magnification). Three mice per group. (<b>C</b>) Representative image of complement C3 deposition. Green signals refer C3 deposition. Scale bar: 300 μm. Six-week-old Fas<sup>lpr</sup> mice were used as a pre-onset control. Results were expressed as mean ± SD. For multiple comparison, the significance was defined as * <span class="html-italic">p</span> &lt; 0.025; ** <span class="html-italic">p</span> &lt; 0.005; *** <span class="html-italic">p</span> &lt; 0.0005 when n = 2. One-way ANOVA Dunnett’s test was used for multiple comparison with 95% confidence interval.</p>
Full article ">Figure 3
<p>Both anti-dsDNA and isotype antibodies were reduced by CL-MSCs treatment: (<b>A</b>) Anti-dsDNA antibody changes in Fas<sup>lpr</sup> mice treated with dPBS (n = 9) and CL-MSCs (n = 10) at three weeks PT. Six-week-old Fas<sup>lpr</sup> mice (n = 12) were used as pre-onset control. (<b>B</b>) Isotype antibody changes in Fas<sup>lpr</sup> mice treated with dPBS (n = 5) and CL-MSCs (n = 6) at three weeks PT. Six-week-old Fas<sup>lpr</sup> mice (n = 8) were used as a pre-onset control. Results were expressed as mean ± SD. For multiple comparisons, the significance was defined as * <span class="html-italic">p</span> &lt; 0.025; ** <span class="html-italic">p</span> &lt; 0.005; *** <span class="html-italic">p</span> &lt; 0.0005; **** <span class="html-italic">p</span> &lt; 0.00005 when n = 2. One-way ANOVA Dunnett’s test was used for multiple comparison with 95% confidence interval.</p>
Full article ">Figure 4
<p>CL-MSCs mediated immunomodulation through targeting monocytes/macrophages and neutrophils and their pro-inflammatory cytokines: (<b>A</b>) The immune profile was analyzed on mouse spleen cells by flow cytometric analysis in six-week-old pre-onset Fas<sup>lpr</sup> mice (n = 7) and at three weeks PT treated with dPBS (n = 7) and CL-MSCs (n = 9). (<b>B</b>) Cytokine profile of plasma samples was performed in six-week-old Fas<sup>lpr</sup> mice (n = 9) and at three weeks PT treated with dPBS (n = 8) and CL-MSCs (n = 10) by Luminex assay. The results are expressed as the mean ± SD. For multiple comparisons, the significance was defined as * <span class="html-italic">p</span> &lt; 0.025; ** <span class="html-italic">p</span> &lt; 0.005; *** <span class="html-italic">p</span> &lt; 0.0005; **** <span class="html-italic">p</span> &lt; 0.00005 when n = 2. One-way ANOVA Dunnett’s test was used for multiple comparison with 95% confidence interval.</p>
Full article ">Figure 5
<p>RNA sequencing confirmed CL-MSCs mediate immunomodulation via the concerted action of proinflammatory cytokine-induced chemokines and production of nitric oxide: (<b>A</b>) Principal component analysis (PCA) comparing 26-week-old Fas<sup>lpr</sup> mice treated with four doses of CL-MSCs (n = 4) and 16-week-old Fas<sup>lpr</sup> mice treated with four doses of dPBS (n = 3). Genes with an adjusted <span class="html-italic">p</span> value for false discovery rate (FDR) less than 0.05 were used for PCA. (<b>B</b>) Graphical summary of Ingenuity Pathway Analysis (IPA). (<b>C</b>) IPA showing the top 10 pathways identified by comparing CL-MSCs-treated mice versus control mice. A positive z-score represents activation of the pathway; a negative z-score represents inhibition of the pathway. (<b>D</b>) The fold change in typical genes involved in the top 10 pathways.</p>
Full article ">
16 pages, 10576 KiB  
Article
DNA Nanoflowers’ Amelioration of Lupus Symptoms in Mice via Blockade of TLR7/9’s Signal
by Jing Wang and Mingzhe Gan
Int. J. Mol. Sci. 2022, 23(24), 16030; https://doi.org/10.3390/ijms232416030 - 16 Dec 2022
Cited by 8 | Viewed by 1820
Abstract
Inhibitory oligodeoxynucleotides (INH-ODN) can exert an immunomodulatory effect to specifically block TLR7 and TLR9 signaling in systemic lupus erythematosus (SLE). To extend the half-life of INH-ODN in vivo, the phosphorothioate backbone, instead of the native phosphodiester, is preferred due to its strong resistance [...] Read more.
Inhibitory oligodeoxynucleotides (INH-ODN) can exert an immunomodulatory effect to specifically block TLR7 and TLR9 signaling in systemic lupus erythematosus (SLE). To extend the half-life of INH-ODN in vivo, the phosphorothioate backbone, instead of the native phosphodiester, is preferred due to its strong resistance against nuclease degradation. However, its incomplete degradation in vivo may lead to potential risk. To solve these problems and enhance the blockage of TLR7 and TLR9, we prepared highly compressed DNA nanoflowers with prolonged native DNA backbones and repeated INH-ODN motifs. Three therapeutic types of nanoflower, incorporating INH-ODN sequences, including IRS 661, IRS 869, and IRS 954, were prepared by rolling circle amplification and were subcutaneously injected into MRL/lpr mice. The TLR7 blocker of the IRS 661 nanoflower and the TLR9 antagonist of the IRS 869 nanoflower could decrease autoantibodies, reduce cytokine secretion, and alleviate lupus nephritis in mice. However, the IRS 954 nanoflower, the TLR7 and TLR9 dual antagonist, did not have additive or opposing effects on lupus nephritis but only showed a decrease in serum IFNα, suggesting that the TLR7 and TLR9 antagonist may have a competition mechanism or signal-dependent switching relationship. INH-ODN nanoflowers were proposed as a novel and potential therapeutic nucleic acids for SLE. Full article
(This article belongs to the Special Issue Design and Synthesis of Biomedical Polymer Materials)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Microphotograph of nanoflower (magnification: left × 20,000, right × 50,000). (<b>A</b>). Prepared IRS 661 DNA nanoflower; (<b>B</b>). prepared IRS 869 DNA nanoflower; (<b>C</b>). prepared IRS 954 DNA nanoflower; (<b>D</b>). prepared CTR DNA nanoflower.</p>
Full article ">Figure 2
<p>Fluorescence images of ANA tests. (<b>A</b>). Model group; (<b>B</b>). CTR-nanoflower-treated group; (<b>C</b>). IRS-661-nanoflower-treated group; (<b>D</b>). IRS-869-nanoflower-treated group; (<b>E</b>). IRS-954-nanoflower-treated group. Scale bar: 50 μm.</p>
Full article ">Figure 3
<p>ANA and anti-dsDNA antibody titers. (<b>a</b>) Serum levels of ANA in serum; (<b>b</b>) serum levels of anti-dsDNA titer in serum. **: Compared with the model group, <span class="html-italic">p</span> &lt; 0.01; ##: compared with the 954-treated group, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Proteinuria value of each group in different weeks.</p>
Full article ">Figure 5
<p>Renal pathological sections examination. (<b>a</b>). Model control group; (<b>b</b>). CTR-nanoflower-treated group; (<b>c</b>). IRS-661-nanoflower-treated group; (<b>d</b>). IRS-869-nanoflower-treated group; (<b>e</b>). IRS-954-nanoflower-treated group; (<b>f</b>). glomerulonephritis score; (<b>g</b>). tubular nephritis score (**: compared with the model control group, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>Renal immune complex examination. (<b>A</b>). Model group; (<b>B</b>). CTR-nanoflower-treated group; (<b>C</b>). IRS-661-nanoflower-treated group; (<b>D</b>). IRS-869-nanoflower-treated group; (<b>E</b>). IRS-954-nanoflower-treated group. Scale bar: 50 μm.</p>
Full article ">Figure 7
<p>Interferon-α and IL-17 contents in mice serum. a: Compared with the model group, <span class="html-italic">p</span> &lt; 0.05; b: compared with the 954-treated group, <span class="html-italic">p</span> &lt; 0.05; c: compared with the model group, <span class="html-italic">p</span> &lt; 0.01; d: compared with the CTR group, <span class="html-italic">p</span> &lt; 0.01; e: compared with the CTR group, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 8
<p>Schematic diagram of the preparation process of DNA nanoflowers.</p>
Full article ">
18 pages, 5604 KiB  
Article
Quercetin Ameliorates Renal Injury and Pyroptosis in Lupus Nephritis through Inhibiting IL-33/ST2 Pathway In Vitro and In Vivo
by Hsin-Yuan Chen, Yi-Fen Chiang, Yong-Han Hong, Tzong-Ming Shieh, Tsui-Chin Huang, Mohamed Ali, Hsin-Yi Chang, Kai-Lee Wang and Shih-Min Hsia
Antioxidants 2022, 11(11), 2238; https://doi.org/10.3390/antiox11112238 - 13 Nov 2022
Cited by 15 | Viewed by 4343
Abstract
Lupus nephritis (LN) is a common and serious symptom in patients with systemic lupus erythematosus (SLE). Tubular interstitial fibrosis is a common underlying mechanism in the development of lupus nephritis to end-stage renal failure (ESRD). Quercetin is widely proven to prevent tissue fibrosis. [...] Read more.
Lupus nephritis (LN) is a common and serious symptom in patients with systemic lupus erythematosus (SLE). Tubular interstitial fibrosis is a common underlying mechanism in the development of lupus nephritis to end-stage renal failure (ESRD). Quercetin is widely proven to prevent tissue fibrosis. Therefore, the purpose of this study is to investigate the beneficial effects of quercetin on the inhibition of fibrosis and inflammation pathways in in vitro and in vivo lupus nephritis models. In the current study, MRL/lpr mice as animal models, and HK-2 human renal tubular epithelial cells were stimulated by interleukin-33 (IL-33) to mimic the cellular model of lupus nephritis. Immunohistochemical staining, immunoblotting assay, immunofluorescence staining, and quantitative real-time polymerase chain reaction assay were used. The in vivo results showed that quercetin improved the renal function and inhibited both fibrosis- and inflammation-related markers in MRL/lpr mice animal models. The in vitro results indicated that quercetin ameliorated the accumulation of fibrosis- and inflammation-related proteins in IL-33-induced HK-2 cells and improved renal cell pyroptosis via the IL33/ST2 pathway. Overall, quercetin can improve LN-related renal fibrosis and inflammation, which may offer an effective potential therapeutic strategy for lupus nephritis. Full article
(This article belongs to the Special Issue Oxidative Stress in Inflammatory Skin and Tissue Disorders)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Quercetin ameliorated the renal pathology in MRL/lpr mice. (<b>A</b>) In vivo experimental procedure. (<b>B</b>) Body weight change in MRL/lpr mice. (<b>C</b>) Changes in appearance and weight of spleen, kidney, and uterus in MRL/lpr mice. (<b>D</b>) Determination of BUN and plasma creatinine concentration. (<b>E</b>) Renal tissue specimen stained with hematoxylin and eosin stain, Sirius Red stain, or Toluidine Blue stain for MRL/lpr mice that were given either vehicle (ddH<sub>2</sub>O) or quercetin for 3 months. Magnification, 40× and 400×. (<b>F</b>) Representative photomicrographs of the expression and distribution of IL-33, NLRP3, and IL-1β in renal tissues in MRL/lpr mice using immunohistochemical staining. Magnification, 40× and 200×. Data were expressed as mean ± SD for each group. * <span class="html-italic">p</span> &lt; 0.05 versus untreated MRL/lpr mice (<span class="html-italic">n</span> = 5).</p>
Full article ">Figure 2
<p>Quercetin reduced fibrosis-related proteins expression in human renal tubular epithelial cell line. HK-2 cells were co-cultured with 5 μM quercetin and 50 ng/mL or 100 ng/mL recombinant human IL-33 for 48 h. (<b>A</b>,<b>B</b>) Representative photomicrographs show the immunofluorescence staining for vimentin and fibronectin among different groups. Magnification, 400×. (<b>C</b>–<b>E</b>) Immunoblots and quantitative histograms showing the results of Western blotting analysis for the expression of vimentin and fibronectin protein. (<b>F</b>–<b>G</b>) Quantitative histograms showing the results of qPCR analysis for <span class="html-italic">VIM</span> and <span class="html-italic">FN1</span> mRNA level. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05 versus control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus corresponding IL-33-stimulated group; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.001 versus corresponding IL-33-stimulated group.</p>
Full article ">Figure 3
<p>Quercetin diminished inflammatory cytokines expression in human renal tubular epithelial cell line. HK-2 cells were co-cultured with 5 μM quercetin and 50 ng/mL or 100 ng/mL recombinant human IL-33 for 48 h. (<b>A</b>–<b>E</b>) Immunoblots and quantitative histograms showing the results of Western blotting analysis for the expression of IL-6, IL-8, IL-33, and HMGB1 proteins. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05 versus control group; ** <span class="html-italic">p</span> &lt; 0.001 versus control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus corresponding IL-33-stimulated group; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.001 versus corresponding IL-33-stimulated group.</p>
Full article ">Figure 4
<p>Quercetin reduced inflammasome-associated proteins expression in human renal tubular epithelial cell line. HK-2 cells were co-cultured with 5 μM quercetin and 50 ng/mL or 100 ng/mL recombinant human IL-33 for 48 h. (<b>A</b>,<b>B</b>) Representative photomicrographs showing the immunofluorescence staining for NLRP3 (red) and ASC (green) among different groups. Boxed areas are enlarged and presented in the right column. Magnification, 400×. (<b>C</b>–<b>F</b>) Immunoblots and quantitative histograms showing the results of Western blotting analysis for the expression of NLRP3, ASC, and caspase-1 proteins. (<b>G</b>) Quantitative histograms showing the results of qPCR analysis for <span class="html-italic">PYCARD</span> mRNA level. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05 versus control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus corresponding IL-33-stimulated group; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.001 versus corresponding IL-33-stimulated group.</p>
Full article ">Figure 5
<p>Quercetin reduced pyroptosis-associated proteins expression in human renal tubular epithelial cell line. HK-2 cells were co-cultured with 5 μM quercetin and 50 ng/mL or 100 ng/mL recombinant human IL-33 for 48 h. (<b>A</b>) Representative photomicrographs showing the immunofluorescence staining for IL-1β (red) among different groups. Magnification, 400×. (<b>B</b>) Quantitative histograms showing the results of qPCR analysis for <span class="html-italic">GSDMD</span> mRNA level. (<b>C</b>–<b>E</b>) Immunoblots and quantitative histograms showing the results of Western blotting analysis for the expression of GSDMD and IL-1β protein. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05 versus control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus corresponding IL-33-stimulated group.</p>
Full article ">Figure 6
<p>Quercetin suppressed the expression of receptor proteins in human renal tubular epithelial cell line. HK-2 cells were co-cultured with 5 μM quercetin and 50 ng/mL or 100 ng/mL recombinant human IL-33 for 48 h. (<b>A</b>–<b>F</b>) Immunoblots and quantitative histograms showing the results of Western blotting analysis for the expression of ST2, IL1RAP, TLR4, TLR7, and TLR9 proteins. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05 versus control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus corresponding IL-33-stimulated group.</p>
Full article ">Figure 7
<p>Quercetin regulated the expression of inflammation-related proteins in human renal tubular epithelial cell line. HK-2 cells were co-cultured with 5 μM quercetin and 50 ng/mL or 100 ng/mL recombinant human IL-33 for 48 h. (<b>A</b>–<b>F</b>) Immunoblots and quantitative histograms showing the results of Western blotting analysis for the expression of NF-κB, MyD88, IκBα, and p-AMPK proteins. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05 versus control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus corresponding IL-33-stimulated group; n.s. indicates not significant.</p>
Full article ">Figure 8
<p>Schematic representation of the mechanisms through which quercetin exerts its potential effects in lupus nephritis-induced renal injury progression. Quercetin suppresses IL-33/ST2 signaling pathway, inhibits inflammasome and pyroptosis-related proteins expression, and reduces the secretion of IL-6 and IL-8 cytokines. Abbreviations: IL-33, interleukin-33; ST2, suppression of tumorigenicity-2; TLR4, toll-like receptors 4; IL-1RAcP, IL-1 receptor accessory protein; MyD88, myeloid differentiation primary response protein 88; IRAK, IL-1R-associated kinase; TRAF6, tumor necrosis factor receptor-associated factor 6; IκBα, inhibitor of nuclear factor κBα; NF-κB, nuclear factor-kappa B; GSDMD, gasdermin D; ASC, apoptosis-associated speck-like protein containing a caspase recruitment domain; NLRP3, NOD-like receptor family pyrin domain-containing protein 3; caspase-1, cysteine aspartate-specific proteases-1.</p>
Full article ">
12 pages, 2169 KiB  
Article
Chlorogenic Acid Relieves the Lupus Erythematosus-like Skin Lesions and Arthritis in MRL/lpr Mice
by Ruxuan Wang, Xiaoyi Yang, Shen You, Mengyao Hao, Jianguang Li, Xiaoguang Chen and Jing Jin
Pharmaceuticals 2022, 15(11), 1327; https://doi.org/10.3390/ph15111327 - 27 Oct 2022
Cited by 7 | Viewed by 2760
Abstract
Chlorogenic acid (CGA) is a phenylpropyl substance synthesized through the shikimic acid pathway. In addition to its anti-tumor, anti-inflammatory, and antioxidant abilities, CGA also has immunomodulatory effects. The aim of the present study is to investigate the therapeutic effects of CGA on the [...] Read more.
Chlorogenic acid (CGA) is a phenylpropyl substance synthesized through the shikimic acid pathway. In addition to its anti-tumor, anti-inflammatory, and antioxidant abilities, CGA also has immunomodulatory effects. The aim of the present study is to investigate the therapeutic effects of CGA on the skin damage and arthritis caused by systemic lupus erythematosus (SLE) in an MRL/lpr mouse model. In the SLE model, female MRL/lpr mice at the age of 10 weeks old were treated with CGA daily or cyclophosphamide (CTX) weekly via intraperitoneal injection for three months. After treatment, CGA can significantly alleviate the skin and mucous membrane damage caused by SLE and has a certain improvement effect on arthritis. CGA could inhibit dsDNA expression to a certain extent but has no obvious regulation on ANA concentration. The ELISA and BioMAP results indicated that CGA might play an anti-inflammatory role by down-regulating the interleukin (IL)-17 level. In conclusion, our study demonstrates that CGA can alleviate multiorgan damage in MRL/lpr mice by reducing IL-17. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Ameliorative effect of chlorogenic acid (CGA) on skin damage in MRL/lpr mice. (<b>A</b>) Schematic of experimental design. One mouse in the model group died at 21 weeks of age while one mouse in the CGA group died at 17 weeks of age (<b>B</b>), which may be related to the disease process. (<b>C</b>) Photos of the entire body of MRL/lpr mice at 22 weeks. Mice in the model group showed skin damage to the back, nose, and mouth. (<b>D</b>) Incidence of skin damage at 3 months. (<b>E</b>) Body weights from 10 to 22 weeks. Control group: <span class="html-italic">n</span> = 8; model group: <span class="html-italic">n</span> = 8; cyclophosphamide (CTX) group: <span class="html-italic">n</span> = 7; CGA group: <span class="html-italic">n</span> = 8. Changes were tested using the chi-squared statistic from SPSS 13.0. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. model.</p>
Full article ">Figure 2
<p>CGA treatment suppressed skin disease in MRL/lpr mice. HE staining (×100) (<b>A</b>) and toluidine blue staining (×100) (<b>B</b>) of the skin tissue treated with CGA and CTX. (<b>C</b>,<b>D</b>) The pathological score of acanthosis and hypertrophy. (<b>E</b>) Mast cells detected by toluidine blue in the affected skin of mice were counted. Control group: <span class="html-italic">n</span> = 8; model group: <span class="html-italic">n</span> = 8; CTX group: <span class="html-italic">n</span> = 7; CGA group: <span class="html-italic">n</span> = 7. The values are expressed as the mean ± SD. Changes were tested using the chi-squared statistic from SPSS 13.0. # <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 vs. control. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 vs. model.</p>
Full article ">Figure 3
<p>CGA treatment alleviated lupus-like arthritis in MRL/lpr mice. We evaluated the effect through front paw volume of left (<b>A</b>) and right (<b>B</b>), HE staining × 100 (<b>C</b>), and pathology score (<b>D</b>). Control group: <span class="html-italic">n</span> = 8; model group: <span class="html-italic">n</span> = 7; CTX group: <span class="html-italic">n</span> = 7; CGA group: <span class="html-italic">n</span> = 8. The values are expressed as the mean ± SD. Data were analyzed using one-way ANOVA test. # <span class="html-italic">p</span> &lt; 0.05, #### <span class="html-italic">p</span> &lt; 0.0001 vs. control, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 vs. model.</p>
Full article ">Figure 4
<p>Changes in SLE-specific indicators of dsDNA (<b>A</b>) and antinuclear antibodies (ANA) (<b>B</b>,<b>C</b>). CTX significantly reduced serum dsDNA levels and ANA titers in a time-dependent manner, while CGA had a tendency to reduce dsDNA but did not significantly change the ANA titers. Fourth week: control group: <span class="html-italic">n</span> = 8; model group: <span class="html-italic">n</span> = 8; CTX group: <span class="html-italic">n</span> = 7; CGA group: <span class="html-italic">n</span> = 8. Eighth and tenth week: control group: <span class="html-italic">n</span> = 8; model group: <span class="html-italic">n</span> = 8; CTX group: <span class="html-italic">n</span> = 7; CGA group: <span class="html-italic">n</span> = 7. Twelfth week: control group: <span class="html-italic">n</span> = 8; model group: <span class="html-italic">n</span> = 7; CTX group: <span class="html-italic">n</span> = 7; CGA group: <span class="html-italic">n</span> = 7. #### <span class="html-italic">p</span> &lt; 0.0001 vs. control, **** <span class="html-italic">p</span> &lt; 0.0001 vs. model. The values are expressed as the mean ± SD. The ordinary one-way ANOVA was used to compare the values between groups.</p>
Full article ">Figure 5
<p>CGA decreases inflammatory cytokines levels in MRL/lpr mice. Serum concentrations of interleukin (IL)-17A, IL-17F, IL-6, and interferon-γ (IFN-γ) were measured in Balb/c and MRL/lpr mice at 22 weeks of age (mean ± SD, <span class="html-italic">n</span> = 7–8/group). Mice were treated with CTX and CGA for 12 weeks. Control group: <span class="html-italic">n</span> = 8; model group: <span class="html-italic">n</span> = 7; CTX group: <span class="html-italic">n</span> = 7; CGA group: <span class="html-italic">n</span> = 7. #### <span class="html-italic">p</span> &lt; 0.0001 vs. control, * <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 vs. model.</p>
Full article ">Figure 6
<p>The biological activity of CGA was tested in 3 BioMAP systems at 4 concentrations (<b>A</b>) and overlays of CGA (50 μM) and methotrexate (MTX) (100 μM) (<b>B</b>). The readout proteins are shown along the <span class="html-italic">x</span>-axis. The grey region closest to the axis represents the range of historical vehicle control data. The annotated peaks constitute biomarker activities that show statistical significance when compared to historical vehicle controls after compound therapy.</p>
Full article ">
15 pages, 2567 KiB  
Article
Cathepsin S Inhibition Suppresses Experimental Systemic Lupus Erythematosus-Associated Pulmonary Arterial Remodeling
by Tzung-Hai Yen, Wan-Jing Ho, Yung-Hsin Yeh and Ying-Ju Lai
Int. J. Mol. Sci. 2022, 23(20), 12316; https://doi.org/10.3390/ijms232012316 - 14 Oct 2022
Cited by 2 | Viewed by 2229
Abstract
Patients with systemic lupus erythematosus (SLE) associated with pulmonary arterial hypnertension (PAH) receive targeted therapy for PAH to decrease pulmonary arterial systolic pressure and significantly prolong their survival. Cysteine cathepsin proteases play critical roles in the progression of cardiovascular disease. Inhibition of cathepsin [...] Read more.
Patients with systemic lupus erythematosus (SLE) associated with pulmonary arterial hypnertension (PAH) receive targeted therapy for PAH to decrease pulmonary arterial systolic pressure and significantly prolong their survival. Cysteine cathepsin proteases play critical roles in the progression of cardiovascular disease. Inhibition of cathepsin S (Cat S) has been shown to improve SLE and lupus nephritis. However, the effect of Cat S inhibitors on SLE-associated PAH (SLE-PAH) remains unclear, and there is no animal model for translational research on SLE-PAH. We hypothesized that the inhibition of Cat S may affect PAH development and arterial remodeling associated with SLE. A female animal model of SLE-PAH, female MRL/lpr (Lupus), was used to evaluate the role of pulmonary arterial remodeling in SLE. The key finding of the research work is the establishment of an animal model of SLE associated with PAH in female MRL/lpr mice that is able to evaluate pulmonary arterial remodeling starting from the age of 11 weeks to 15 weeks. Cat S protein level was identified as a marker of experimental SLE. Pulmonary hypertension in female MRL/lpr (Lupus) mice was treated by administering the selective Cat S inhibitor Millipore-219393, which stimulated peroxisome proliferator-activated receptor-gamma (PPARγ) in the lungs to inhibit Cat S expression and pulmonary arterial remodeling. Studies provide an animal model of female MRL/lpr (Lupus) associated with PAH and a deeper understanding of the pathogenesis of SLE-PAH. The results may define the role of cathepsin S in preventing progressive and fatal SLE-PAH and provide approaches for therapeutic interventions in SLE-PAH. Full article
Show Figures

Figure 1

Figure 1
<p>Increased cathepsin S and decreased PPARγ expression are the markers of PAH related to SLE. (<b>A</b>) Western blot analysis of cathepsin S and PPARγ in the organs of female MRL/lpr mice (SLE) compared to male MRL/lpr (Con) mice. (<b>B</b>,<b>C</b>) Relative expression values obtained by densitometry of the cathepsin S and PPARγ protein normalized to GAPDH (<span class="html-italic">n</span> = 3 per group). GAPDH = glyceraldehyde-3-phosphate dehydrogenase. The data are presented as the mean ± SEM of four samples in each group. * <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 compared with the control (Con) group. (<b>D</b>). Representative RVSP tracings of male and female MRL/lpr mice at the age of 15 weeks. Pulmonary hypertension (indicated by an elevated RVSP) showed increased RVSPs in the female MRL/lpr mice (SLE) groups. Increased cathepsin S and loss PPARγ protein expression enhanced the RVSP in female MRL/lpr mice (SLE). The data are presented as the mean ± SEM of four samples in each group. *** <span class="html-italic">p</span> &lt; 0.001 compared with the Con group.</p>
Full article ">Figure 1 Cont.
<p>Increased cathepsin S and decreased PPARγ expression are the markers of PAH related to SLE. (<b>A</b>) Western blot analysis of cathepsin S and PPARγ in the organs of female MRL/lpr mice (SLE) compared to male MRL/lpr (Con) mice. (<b>B</b>,<b>C</b>) Relative expression values obtained by densitometry of the cathepsin S and PPARγ protein normalized to GAPDH (<span class="html-italic">n</span> = 3 per group). GAPDH = glyceraldehyde-3-phosphate dehydrogenase. The data are presented as the mean ± SEM of four samples in each group. * <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 compared with the control (Con) group. (<b>D</b>). Representative RVSP tracings of male and female MRL/lpr mice at the age of 15 weeks. Pulmonary hypertension (indicated by an elevated RVSP) showed increased RVSPs in the female MRL/lpr mice (SLE) groups. Increased cathepsin S and loss PPARγ protein expression enhanced the RVSP in female MRL/lpr mice (SLE). The data are presented as the mean ± SEM of four samples in each group. *** <span class="html-italic">p</span> &lt; 0.001 compared with the Con group.</p>
Full article ">Figure 2
<p>Female MRL/lpr mice showed increased development of pulmonary arterial remodeling and right ventricle hypertrophy. (<b>A</b>) Media wall thickness (MWT) values of small PAs (50–100 µm) identified by SMA staining. The data are presented as the mean ± SEM (<span class="html-italic">n</span> = 3–5). *** <span class="html-italic">p</span> &lt; 0.001 versus male MRL/lpr mice (Con); one-way ANOVA. (<b>B</b>) The ratios of RV to LV plus the septum weight (RV/LV+S) in 5 different groups are shown for male MRL/lpr mice and female MRL/lpr mice. The data are presented as the mean ± SEM (<span class="html-italic">n</span> = 3–5), * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with male MRL/lpr mice. one-way ANOVA.</p>
Full article ">Figure 3
<p>The degree of muscularization of pulmonary arteries in male and female MRL/lpr mice. (<b>A</b>) Degree of non (N), partial (P), or full (M) muscularization. (<b>B</b>) Percentage of non (N)-, partially (P)-, or fully (M)-muscularized pulmonary arteries, as a percentage of total pulmonary arteries (size 25–100 µm) in a cross section. A total of 60–80 intra-acinar vessels was analyzed in each lung. The results from 11-week and 15-week groups (Con mice and SLE mice) are presented. The data are presented as the mean ± SE. *** <span class="html-italic">p</span> &lt;0.001 versus the Con group; ### <span class="html-italic">p</span> &lt; 0.001 versus the Con group, showed a significantly different increase or decrease from the Con group. §§ <span class="html-italic">p</span> &lt; 0.01 versus the 11-week SLE group. ANOVA with Bonferroni’s post hoc test.</p>
Full article ">Figure 4
<p>Effect of Cat S inhibitor on PPARγ stimulation, and decreasing Cat S expression in the lungs and circulation of female MRL/lpr mice. (<b>A</b>) Representative immunoblotting densitometry quantification of protein expression is indicated for the lungs of the three groups. Cat S (24 kDa band) and PPARγ (57 kDa band). The bars represent the mean ± SEM for <span class="html-italic">n</span> = 3 samples. * <span class="html-italic">p</span> &lt; 0.05 (increase) or ## <span class="html-italic">p</span> &lt; 0.05 (decrease) compared with the male MRL/lpr mice (Con) group. (<b>B</b>) Cat S activity in the lung tissue lysate. (<b>C</b>) Cat S activity in the serum. Each value (mean ± SE (<span class="html-italic">n</span> = 6–7) is expressed. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 (increase) versus male MRL/lpr (Con) mice, or # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 (decrease) compared with the female MRL/lpr mice, one-way ANOVA, Bonferroni posttest.</p>
Full article ">Figure 5
<p>Effect of Cat S inhibitor in preventing the development of pulmonary arterial remodeling and right ventricle hypertrophy. (<b>A</b>) Immunohistochemistry analysis of SMA with cathepsin S, and (<b>B</b>) SMA with PPARγ in the pulmonary vascularity (scale bar: 25 µm). (<b>C</b>) Medial wall thickness (MWT) of small PAs identified by SMA staining. The degree of MWT was compared among 3 groups. (<b>D</b>) The ratio of RV to LV plus septum weight (RV/LV+S) is shown. Each value (mean ± SEM (<span class="html-italic">n</span> = 3–5)) is expressed. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 versus male MRL/lpr mice (Con) group or ### <span class="html-italic">p</span> &lt; 0.001 compared with the female MRL/lpr mice (SLE) group, one-way ANOVA, Bonferroni posttest.</p>
Full article ">
18 pages, 3257 KiB  
Article
Lipidomics Revealed Aberrant Lipid Metabolism Caused by Inflammation in Cardiac Tissue in the Early Stage of Systemic Lupus Erythematosus in a Murine Model
by Jida Zhang, Lu Lu, Xiaoyu Tian, Kaili Wang, Guanqun Xie, Haichang Li, Chengping Wen and Changfeng Hu
Metabolites 2022, 12(5), 415; https://doi.org/10.3390/metabo12050415 - 5 May 2022
Cited by 8 | Viewed by 2139
Abstract
Cardiac involvement, displayed as premature cardiovascular disease (CVD), is one of common clinical symptoms of patients with systemic lupus erythematosus (SLE), contributing to mortality of the disease. The precise underlying pathological mechanism(s) for the cardiac involvement in lupus remains poorly understood. Lipids and [...] Read more.
Cardiac involvement, displayed as premature cardiovascular disease (CVD), is one of common clinical symptoms of patients with systemic lupus erythematosus (SLE), contributing to mortality of the disease. The precise underlying pathological mechanism(s) for the cardiac involvement in lupus remains poorly understood. Lipids and their metabolites are directly involved in atherosclerosis development, oxidative stress, and inflammation, which are closely related to the development of CVD. In the study, shotgun lipidomics was exploited to quantitatively analyze cellular lipidomes in the cardiac tissue of MRL/lpr mice at two different time points (i.e., pre-lupus and lupus state) with/without treatment with glucocorticoids (GCs). Urine protein, spleen index, and renal histopathological evaluation of the mice were also performed for assessment of SLE onset and/or outcome. Lipidomics analysis revealed that the deposition of cholesterol and the aberrant metabolism of lipids caused by the increased energy metabolism and the enhanced activation of phospholipases, both of which were originally induced by inflammation, were already present in cardiac tissues from lupus-prone mice even at pre-lupus state. These lipid alterations could further induce inflammation and autoimmune responses, accelerating the process of CVD. In addition, the present study also demonstrated that GCs therapy could not only delay the progression of SLE, but also partially corrected these alterations of lipid species in cardiac tissue due to their anti-inflammatory effect. Thus, the medications with better anti-inflammatory effect might be a useful therapeutic method for premature CVD of SLE. Full article
Show Figures

Figure 1

Figure 1
<p>Representative images of renal histopathology of MRL/lpr mice from each group at two different time points. Renal tissues of the control (<span class="html-italic">n</span> = 4), model (<span class="html-italic">n</span> = 4), and glucocorticoids (GCs) (<span class="html-italic">n</span> = 4) groups were collected at 8 and 14 weeks of age, respectively. Both model and GCs groups were MRL/lpr mice, while the control group was MRL/MpJ mice. Kidney pathological changes were determined by H&amp;E staining (Panel <b>A</b>). Images were captured under ×100 visual field with optical microscopy. IgG deposition in glomeruli was assessed through immunofluorescence analysis (nucleus stained with 4′, 6-diamidino-2-phenylindole, blue; IgG antibody, green) (Panel <b>B</b>). Images were obtained under ×400 visual field with fluorescence microscopy. The C3 deposition in glomeruli as assessed by immunohistochemical staining (Panel <b>C</b>). the representative images were captured under ×100 visual field. The intensities of IgG (Panel <b>D</b>) and C3 deposition (Panel <b>E</b>) on the glomeruli from two to four sections from each mouse were determined and displayed as normalized values of the model group. Difference between the groups was determined using ANOVA followed by Dunn multiple comparison with IBM SPSS Statistics 19 Software (SPSS Inc., Chicago, IL, USA) after the normality of each group of variables was checked by Q–Q plots. The raw <span class="html-italic">p</span> values were adjusted to false discovery rat using the Benjamini–Hochberg method. *** adjusted <span class="html-italic">p</span> &lt; 0.001 and <sup>###</sup> adjusted <span class="html-italic">p</span> &lt; 0.001 compared with those in the control and the model group, respectively. H&amp;E represents hematoxylin and eosin, and C3 denotes Complement C3.</p>
Full article ">Figure 1 Cont.
<p>Representative images of renal histopathology of MRL/lpr mice from each group at two different time points. Renal tissues of the control (<span class="html-italic">n</span> = 4), model (<span class="html-italic">n</span> = 4), and glucocorticoids (GCs) (<span class="html-italic">n</span> = 4) groups were collected at 8 and 14 weeks of age, respectively. Both model and GCs groups were MRL/lpr mice, while the control group was MRL/MpJ mice. Kidney pathological changes were determined by H&amp;E staining (Panel <b>A</b>). Images were captured under ×100 visual field with optical microscopy. IgG deposition in glomeruli was assessed through immunofluorescence analysis (nucleus stained with 4′, 6-diamidino-2-phenylindole, blue; IgG antibody, green) (Panel <b>B</b>). Images were obtained under ×400 visual field with fluorescence microscopy. The C3 deposition in glomeruli as assessed by immunohistochemical staining (Panel <b>C</b>). the representative images were captured under ×100 visual field. The intensities of IgG (Panel <b>D</b>) and C3 deposition (Panel <b>E</b>) on the glomeruli from two to four sections from each mouse were determined and displayed as normalized values of the model group. Difference between the groups was determined using ANOVA followed by Dunn multiple comparison with IBM SPSS Statistics 19 Software (SPSS Inc., Chicago, IL, USA) after the normality of each group of variables was checked by Q–Q plots. The raw <span class="html-italic">p</span> values were adjusted to false discovery rat using the Benjamini–Hochberg method. *** adjusted <span class="html-italic">p</span> &lt; 0.001 and <sup>###</sup> adjusted <span class="html-italic">p</span> &lt; 0.001 compared with those in the control and the model group, respectively. H&amp;E represents hematoxylin and eosin, and C3 denotes Complement C3.</p>
Full article ">Figure 2
<p>Comparison of the manifestations of MRL/lpr mice from each group at different states. Fresh urine samples and spleen tissues of the control (<span class="html-italic">n</span> = 4), model (<span class="html-italic">n</span> = 4), and GCs (<span class="html-italic">n</span> = 4) groups were collected at 8 and 14 weeks of age, respectively. Both model and GCs group were MRL/lpr mice, while the control group was MRL/MpJ mice. (<b>A</b>) Urinary protein levels. (<b>B</b>) The ratio of spleen-to-mouse body weight. The data represent means ± SEM from different groups. Difference between the groups was determined using ANOVA followed by Dunn multiple comparison with IBM SPSS Statistics 19 Software (SPSS Inc., Chicago, IL, USA) after the normality of each group of variables was checked by Q–Q plots. The raw <span class="html-italic">p</span> values were adjusted to false discovery rat using the Benjamini–Hochberg method. * adjusted <span class="html-italic">p</span> &lt; 0.05 and *** adjusted <span class="html-italic">p</span> &lt; 0.001 compared with those in the control group. <sup>#</sup> adjusted <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> adjusted <span class="html-italic">p</span> &lt; 0.01 compared with those in the model group. <span class="html-italic">NS</span>, not significant.</p>
Full article ">Figure 3
<p>Comparison of the total amount of triacylglycerol species and cholesterol deposited in cardiac tissues from MRL/lpr mice from each group at different states. Heart tissues of the control (<span class="html-italic">n</span> = 4), model (<span class="html-italic">n</span> = 4), and GCs (<span class="html-italic">n</span> = 4) groups were collected at 8 and 14 weeks of age, respectively. Both model and GCs groups were MRL/lpr mice, while the control group was MRL/MpJ mice. Lipidomics analysis of total level of triacylglycerol (TAG) species (Panels <b>A</b> and <b>B</b>), composition of fatty acyls in TAGs (Panel <b>E</b>), and cholesterol level (Panels <b>C</b> and <b>D</b>) present in lipid extracts of heart was performed through multidimensional mass-spectrometry-based shotgun lipidomics. The data represent means ± SEM from different groups. Difference between the groups was determined using ANOVA followed by Dunn multiple comparison with IBM SPSS Statistics 19 Software (SPSS Inc., Chicago, IL, USA) after the normality of each group of variables was checked by Q–Q plots. The raw <span class="html-italic">p</span> values were adjusted to false discovery rat using the Benjamini–Hochberg method. * adjusted <span class="html-italic">p</span> &lt; 0.05 and <sup>#</sup> adjusted <span class="html-italic">p</span> &lt; 0.05 compared with those in the control group and the model group, respectively. <span class="html-italic">NS</span>, not significant.</p>
Full article ">Figure 4
<p>Comparison of total amounts of 4-hydroxyalkenals and cardiolipin species in cardiac tissues from MRL/lpr mice from each group at different states. Heart tissues of the control (<span class="html-italic">n</span> = 4), model (<span class="html-italic">n</span> = 4), and GCs (<span class="html-italic">n</span> = 4) groups were collected at 8 and 14 weeks of age, respectively. Both model and GCs groups were MRL/lpr mice, while the control group was MRL/MpJ mice. Lipidomics analysis of 4-hydroxyalkenal (HNE) species (Panels <b>A</b> and <b>B</b>) and cardiolipin species (Panels <b>C</b>–<b>E</b>) present in lipid extracts of heart was performed through multidimensional mass spectrometry-based shotgun lipidomics. The data represent means ± SEM from different groups. Difference between the groups was determined using ANOVA followed by Dunn multiple comparison with IBM SPSS Statistics 19 Software (SPSS Inc., Chicago, IL, USA) after the normality of each group of variables was checked by Q–Q plots. The raw <span class="html-italic">p</span> values were adjusted to false discovery rat using the Benjamini–Hochberg method. * adjusted <span class="html-italic">p</span> &lt; 0.05, ** adjusted <span class="html-italic">p</span> &lt; 0.01, and *** adjusted <span class="html-italic">p</span> &lt; 0.001 compared with those in the control group. <sup>#</sup> adjusted <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> adjusted <span class="html-italic">p</span> &lt; 0.01 compared with those in the model group. <span class="html-italic">NS</span>, not significant.</p>
Full article ">Figure 5
<p>Comparison of the total amounts of lysophospholipid and phospholipid species in heart tissues from MRL/lpr mice from each group at different states. Heart tissues of the control (<span class="html-italic">n</span> = 4), model (<span class="html-italic">n</span> = 4), and GCs (<span class="html-italic">n</span> = 4) groups were collected at 8 and 14 weeks of age, respectively. Both model and GCs groups were MRL/lpr mice, while the control group was MRL/MpJ mice. Lipidomics analysis of lysophospholipid species, including choline lysoglycerophospholipid (lysoPC) and ethanolamine lysoglycerophospholipid (lysoPE) (Panels <b>A</b>–<b>D</b>), and phospholipid species, i.e., choline glycerophospholipid (PC) and ethanolamine glycerophospholipid (PE) (Panels <b>E</b> and <b>F</b>), present in lipid extracts of heart was performed through multidimensional mass-spectrometry-based shotgun lipidomics. The data represent means ± SEM from different groups. Difference between the groups was determined using ANOVA followed by Dunn multiple comparison with IBM SPSS Statistics 19 Software (SPSS Inc., Chicago, IL., USA) after the normality of each group of variables was checked by Q–Q plots. The raw <span class="html-italic">p</span> values were adjusted to false discovery rat using the Benjamini–Hochberg method. ** adjusted <span class="html-italic">p</span> &lt; 0.01 and *** adjusted <span class="html-italic">p</span> &lt; 0.001 compared with those in the control group. <sup>#</sup> adjusted <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> adjusted <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> adjusted <span class="html-italic">p</span> &lt; 0.001compared with those in the model group. <span class="html-italic">NS</span>, not significant.</p>
Full article ">Figure 6
<p>Comparison of the levels of other classes of phospholipids and sphingomyelin species in heart tissues from MRL/lpr mice from each group. Heart tissues of the control (<span class="html-italic">n</span> = 4), model (<span class="html-italic">n</span> = 4), and GCs (<span class="html-italic">n</span> = 4) groups were collected at 8 weeks of age. Both model and GCs group were MRL/lpr mice, while the control group was MRL/MpJ mice. Lipidomics analysis of other classes of phospholipids, including phosphatidylglycerol (PG) and phosphatidylserine (PS) species (Panels <b>A</b> and <b>B</b>) and sphingomyelin (SM) species (Panel <b>C</b>) present in lipid extracts of heart was performed through multidimensional mass-spectrometry-based shotgun lipidomics. The data represent means ± SEM from different groups. Difference between the groups was determined using ANOVA followed by Dunn multiple comparison with IBM SPSS Statistics 19 Software (SPSS Inc., Chicago, IL, USA) after the normality of each group of variables was checked by Q–Q plots. The raw <span class="html-italic">p</span> values were adjusted to false discovery rat using the Benjamini–Hochberg method. * adjusted <span class="html-italic">p</span> &lt; 0.05 and <sup>#</sup> adjusted <span class="html-italic">p</span> &lt; 0.05 compared with those in the control group and the model group, respectively. <span class="html-italic">NS</span>, not significant.</p>
Full article ">
18 pages, 4623 KiB  
Article
The Ameliorative Effect of Dexamethasone on the Development of Autoimmune Lung Injury and Mediastinal Fat-Associated Lymphoid Clusters in an Autoimmune Disease Mouse Model
by Yaser Hosny Ali Elewa, Md Abdul Masum, Sherif Kh. A. Mohamed, Md Rashedul Islam, Teppei Nakamura, Osamu Ichii and Yasuhiro Kon
Int. J. Mol. Sci. 2022, 23(8), 4449; https://doi.org/10.3390/ijms23084449 - 18 Apr 2022
Cited by 2 | Viewed by 2547
Abstract
In our previous study, we revealed the ameliorative therapeutic effect of dexamethasone (Dex) for Lupus nephritis lesions in the MRL/MpJ-Fas lpr/lpr (Lpr) mouse model. The female Lpr mice developed a greater number of mediastinal fat-associated lymphoid clusters (MFALCs) and inflammatory lung lesions [...] Read more.
In our previous study, we revealed the ameliorative therapeutic effect of dexamethasone (Dex) for Lupus nephritis lesions in the MRL/MpJ-Fas lpr/lpr (Lpr) mouse model. The female Lpr mice developed a greater number of mediastinal fat-associated lymphoid clusters (MFALCs) and inflammatory lung lesions compared to the male mice. However, the effect of Dex, an immunosuppressive drug, on both lung lesions and the development of MFALCs in Lpr mice has not been identified yet. Therefore, in this study, we compared the development of lung lesions and MFALCs in female Lpr mice that received either saline (saline group “SG”) or dexamethasone (dexamethasone group “DG”) in drinking water as a daily dose along with weekly intraperitoneal injections for 10 weeks. Compared to the SG group, the DG group showed a significant reduction in the levels of serum anti-dsDNA antibodies, the size of MFALCs, the degree of lung injury, the area of high endothelial venules (HEVs), and the number of proliferating and immune cells in both MFALCs and the lungs. A significant positive correlation was observed between the size of MFALCs and the cellular aggregation in the lungs of Lpr mice. Therefore, this study confirmed the ameliorative effect of Dex on the development of lung injury and MFALCs via their regressive effect on both immune cells’ proliferative activity and the development of HEVs. Furthermore, the reprogramming of MFALCs by targeting immune cells and HEVs may provide a therapeutic strategy for autoimmune-disease-associated lung injury. Full article
Show Figures

Figure 1

Figure 1
<p>Analysis of the effect of dexamethasone on the degree of lung injury, development of mediastinal fat associated-lymphoid clusters (MFALCs), and autoimmunity in female Lpr mice. (<b>A</b>,<b>B</b>) HE staining of the lung (<b>A</b>) and mediastinal fat tissues “MFTs” (<b>B</b>) in saline and dexamethasone administered groups. Notice the mononuclear cellular aggregations “MNCA” (black arrows) and MFALCs (blue arrows) associated with the MFTs. (<b>C</b>,<b>D</b>) Graphs indicating the morphometrical data of the percentages for the ratios of the area of MNCA/area of the lung field (mm<sup>2</sup>) and the area of LCs/the total area of MFTs (mm<sup>2</sup>). (<b>E</b>) Graph indicating the serum levels of anti-double-stranded DNA autoantibodies. Highly significant values (**) were observed between the saline and dexamethasone groups (<span class="html-italic">p</span> &lt; 0.01), where <span class="html-italic">n</span> = 5 in each experimental group. Analysis was conducted using the Mann–Whitney <span class="html-italic">U</span> test. Data are presented as mean values ± SE.</p>
Full article ">Figure 2
<p>Morphometrical analysis of the effect of dexamethasone on immune cell populations in the lungs and mediastinal fat associated-lymphoid clusters (MFALCs) in female Lpr mice. (<b>A</b>,<b>B</b>) Representative graphs showing the percentage of the positive index ratios for CD3<sup>+</sup> T-lymphocytes (<a href="#app1-ijms-23-04449" class="html-app">Supplementary Figure S1A</a>), B220<sup>+</sup> B-lymphocytes (<a href="#app1-ijms-23-04449" class="html-app">Supplementary Figure S1B</a>), Iba-1<sup>+</sup> macrophages (<a href="#app1-ijms-23-04449" class="html-app">Supplementary Figure S1C</a>), and Gr-1<sup>+</sup> granulocytes (<a href="#app1-ijms-23-04449" class="html-app">Supplementary Figure S1D</a>) in the lungs (<b>A</b>) and MFALCs (<b>B</b>). Significant (*) and highly significant values (**) were observed between the saline and dexamethasone groups (<span class="html-italic">p</span>  values &lt; 0.05, and &lt;0.01, respectively), where <span class="html-italic">n</span> = 5 in each experimental group. Analysis was conducted using the Mann–Whitney <span class="html-italic">U</span> test. Data are presented as mean values ± SE.</p>
Full article ">Figure 3
<p>Analysis of the degree of proliferation of immune cells in the lungs and mediastinal fat associated-lymphoid clusters (MFALCs) in female Lpr mice. (<b>A</b>,<b>C</b>) Representative images of the immunohistochemical-stained lung (<b>A</b>) and mediastinal fat tissue sections (<b>C</b>) stained with anti-Ki67 antibody in both the saline and dexamethasone groups. (<b>B</b>,<b>D</b>) Representative graphs showing the percentage for the ratio of Ki67<sup>+</sup> proliferating cell density in the lungs (<b>B</b>) and Ki67<sup>+</sup> proliferating cell number/total immune cell number in the MFALCs (<b>D</b>). A highly significant value (**) was observed between the saline and dexamethasone groups (<span class="html-italic">p</span> values &lt; 0.01), where <span class="html-italic">n</span> = 5 in each experimental group. Analysis was conducted using the Mann–Whitney <span class="html-italic">U</span> test. Data are presented as mean values ± SE.</p>
Full article ">Figure 4
<p>Analysis of the effect of dexamethasone on the degree of the development of HEVs in the lungs and mediastinal fat associated-lymphoid clusters (MFALCs) in female Lpr mice. (<b>A</b>,<b>B</b>) Representative images of the immunohistochemical-stained lung (<b>A</b>) and mediastinal fat tissue sections (<b>B</b>) stained with anti-PNAd antibody in both the saline and dexamethasone groups. Notice PNAd<sup>+</sup> HEVs (arrows). (<b>C</b>,<b>D</b>) Representative graphs showing the percentage of the relative area ratio of PNAd<sup>+</sup> HEVs in the lungs (<b>C</b>) and MFALCs (<b>D</b>). A highly significant value (**) was observed between saline and dexamethasone groups (<span class="html-italic">p</span> values &lt; 0.01), where <span class="html-italic">n</span> = 5 in each experimental group. Analysis was conducted using the Mann–Whitney <span class="html-italic">U</span> test. Data are presented as mean values ± SE.</p>
Full article ">Figure 5
<p>Analysis of the effect of dexamethasone on the degree of the development of lymphatic vessels (LVs) in the lungs and mediastinal fat associated-lymphoid clusters (MFALCs) in female Lpr mice. (<b>A</b>) Representative images of the dual immunofluorescent stained lung section with anti- CD68<sup>+</sup> macrophages (red) and anti- LYVE-1 (white) antibodies. Notice CD68<sup>+</sup> macrophages (red arrows), LYVE-1<sup>+</sup> LVs (white arrows), and engorged LVs with immune cells (*). (<b>B</b>,<b>C</b>) Representative graphs showing the percentages of LYVE-1<sup>+</sup> LV relative area ratios in the immunohistochemical-stained lungs (<b>B</b>) and MFALCs (<b>C</b>) tissue sections. Highly significant values (**) were observed between the saline and dexamethasone groups (<span class="html-italic">p</span> values &lt; 0.01), where <span class="html-italic">n</span> = 5 in each experimental group. Analysis was conducted using the Mann–Whitney <span class="html-italic">U</span> test. Data are presented as mean values ± SE.</p>
Full article ">Figure 6
<p>Analysis of the effect of dexamethasone on the expression of chemokine in the lungs of female Lpr mice. (<b>A</b>,<b>B</b>) Representative merged images of double immunofluorescent staining for CXCR5 “red” and CD79a “green” positive cells, and Hoechst nuclear staining “blue” (<b>A</b>) along with CXCL13 “green” and B220 “blue” positive cells, and Hoechst nuclear staining “white” (<b>B</b>). Notice positive reactions for CXCR5 (red arrows), B-lymphocytes (blue arrows), CXCL13 (violet arrows), and CXCR5<sup>+/</sup>CD79a<sup>+</sup> co-stained cells (yellow arrows). (<b>C</b>,<b>D</b>) Representative graphs showing the percentage for the ratio of positive cell density for the CXCR5 (<b>C</b>) and CXCL13 chemokines (<b>D</b>). A highly significant value (**) was observed between saline and dexamethasone groups (<span class="html-italic">p</span> values &lt; 0.01), where <span class="html-italic">n</span> = 5 in each experimental group. Analysis was conducted using the Mann–Whitney <span class="html-italic">U</span> test. Data are presented as mean values ± SE.</p>
Full article ">Figure 7
<p>Analysis of the correlations among histoplanimetrical measurements of the lungs and mediastinal fat associated-lymphoid clusters (MFALCs) in the female Lpr mice. (<b>A</b>,<b>B</b>) Representative graphs showing Spearman’s correlations among the size of MFALCs with mononuclear cell aggregates in the lung (<b>A</b>) and HEVs in the MFALCs (<b>B</b>). (<b>C</b>,<b>D</b>) Representative graphs showing Spearman’s correlations among the lung HEVs with MFALC HEVs (<b>C</b>) and mononuclear cell aggregation in the lungs (<b>D</b>); The data were analyzed by the Spearman’s correlation test, where <span class="html-italic">n</span> = 10, <span class="html-italic">ρ</span>: Spearman’s rank-order correlation coefficient. * Significant, <span class="html-italic">p</span> &lt; 0.05, ** Highly significant, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 8
<p>Analysis of the correlations between the immune cell populations and the proliferating cells in the lungs and mediastinal fat associated-lymphoid clusters (MFALCs) in saline and dexamethasone group mice. (<b>A</b>–<b>E</b>) Representative graphs showing Spearman’s correlation between the immune cell populations of the lungs and MFALCs (<b>A</b>), B-lymphocyte populations of the lungs and MFALCs (<b>B</b>), macrophage populations of the lungs and MFALCs (<b>C</b>), granulocyte populations of the lungs and MFALCs (<b>D</b>), proliferating cell populations of the lungs and MFALCs (<b>E</b>). The data were analyzed by Spearman’s correlation test, where <span class="html-italic">n</span> = 10, <span class="html-italic">ρ</span>: Spearman’s rank-order correlation coefficient. * Significant, <span class="html-italic">p</span> &lt; 0.05, ** Highly significant, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
13 pages, 4342 KiB  
Article
Toll-like Receptor Signaling Inhibitory Peptide Improves Inflammation in Animal Model and Human Systemic Lupus Erythematosus
by Wook-Young Baek, Yang-Seon Choi, Sang-Won Lee, In-Ok Son, Ki-Woong Jeon, Sang-Dun Choi and Chang-Hee Suh
Int. J. Mol. Sci. 2021, 22(23), 12764; https://doi.org/10.3390/ijms222312764 - 25 Nov 2021
Cited by 8 | Viewed by 3386
Abstract
Toll-like receptors (TLRs) play a major role in the innate immune system. Several studies have shown the regulatory effects of TLR-mediated pathways on immune and inflammatory diseases. Dysregulated functions of TLRs within the endosomal compartment, including TLR7/9 trafficking, may cause systemic lupus erythematosus [...] Read more.
Toll-like receptors (TLRs) play a major role in the innate immune system. Several studies have shown the regulatory effects of TLR-mediated pathways on immune and inflammatory diseases. Dysregulated functions of TLRs within the endosomal compartment, including TLR7/9 trafficking, may cause systemic lupus erythematosus (SLE). TLR signaling pathways are fine-tuned by Toll/interleukin-1 receptor (TIR) domain-containing adapters, leading to interferon (IFN)-α production. This study describes a TLR inhibitor peptide 1 (TIP1) that primarily suppresses the downstream signaling mediated by TIR domain-containing adapters in an animal model of lupus and patients with SLE. The expression of most downstream proteins of the TLR7/9/myeloid differentiation factor 88 (MyD88)/IFN regulatory factor 7 signaling was downregulated in major tissues such as the kidney, spleen, and lymph nodes of treated mice. Furthermore, the pathological analysis of the kidney tissue confirmed that TIP1 could improve inflammation in MRL/lpr mice. TIP1 treatment downregulated many downstream proteins associated with TLR signaling, such as MyD88, interleukin-1 receptor-associated kinase, tumor necrosis factor receptor-associated factor 6, and IFN-α, in the peripheral blood mononuclear cells of patients with SLE. In conclusion, our data suggest that TIP1 can serve as a potential candidate for the treatment of SLE. Full article
Show Figures

Figure 1

Figure 1
<p>The dramatic inhibitory effect of Toll-like receptor inhibitor peptide 1 (TIP1) on systemic lupus erythematosus (SLE) in a mouse model. (<b>a</b>) Summary of the experimental validation of the inhibitory effect of TIP1 on SLE in a lupus-prone mouse. (<b>b</b>) Images of the whole body of female lupus-prone mice (MRL/<span class="html-italic">lpr</span>) and age- and sex-matched controls (C57BL/6J). Ameliorative effects of TIP1 on inguinal lymphoproliferation (<b>c</b>), nephromegaly (<b>d</b>), and splenomegaly (<b>e</b>). PBS, phosphate-buffered saline. n = 5 mice/group.</p>
Full article ">Figure 2
<p>(<b>a</b>) Anti-dsDNA antibodies, (<b>b</b>) anti-nuclear antibodies (ANA), (<b>c</b>) C3 complement levels in the serum, and (<b>d</b>) albumin content in the urine as determined by enzyme-linked immunosorbent assays (ELISAs). All experiments were performed in duplicate wells (n = 4–5 mice/group). The exact Mann–Whitney U test was performed to compare the mean values between groups. * <span class="html-italic">p</span> &lt; 0.05. TIP1, Toll-like receptor inhibitor peptide 1.</p>
Full article ">Figure 3
<p>Toll-like receptor inhibitor peptide 1 (TIP1) improved nephritis in lupus-prone mice. (<b>a</b>) Periodic acid–Schiff (PAS) staining in the kidneys treated with TIP1. Representative microphotographs from PAS staining of the kidney tissues showing mesangial cell population, endothelial cell proliferation, and inflammatory cell infiltration around the glomerulus of lupus-prone mice (MRL/<span class="html-italic">lpr</span>; left panel, vehicle group; right panel, TIP1-treated group). n = 5 mice/group. (<b>b</b>) The total expression levels and (<b>c</b>) quantitative densitometry of endosomal Toll-like receptor signaling proteins in kidney tissues (n = 4 mice/group). * <span class="html-italic">p</span> &lt; 0.05 by Mann–Whitney <span class="html-italic">U</span> test. The experiments were repeated at least twice. MyD88, myeloid differentiation factor 88; IRAK4, interleukin-1 receptor-associated kinase 4; TRAF3, tumor necrosis factor receptor-associated factor 3; TRAF6, tumor necrosis factor receptor-associated factor 6.</p>
Full article ">Figure 4
<p>Toll-like receptor inhibitor peptide 1 (TIP1) blocks the endosomal Toll-like receptor 7 (TLR7)/TLR9 signaling pathway in the spleen. (<b>a</b>) The expression of the proteins of the endosomal TLR signaling was analyzed by Western blotting using spleen tissues from female lupus-prone mice (MRL/<span class="html-italic">lpr</span>). (<b>b</b>) The summarized bar graph shows the band intensity presented as the ratio of the target protein to actin (n = 4 mice/group). Values are means ± standard deviation (SD). * <span class="html-italic">p</span> &lt; 0.05 by Mann–Whitney <span class="html-italic">U</span> test. (<b>c</b>) Immunohistochemistry was performed to detect endosomal TLR pathway proteins (brown) in the spleen (magnification, ×20). The experiments were repeated at least twice. MyD88, myeloid differentiation factor 88; IRAK4, interleukin-1 receptor-associated kinase 4; TRAF3, tumor necrosis factor receptor-associated factor 3; TRAF6, tumor necrosis factor receptor-associated factor 6.</p>
Full article ">Figure 5
<p>Inhibition of Toll-like receptor 7(TLR7)/TLR9 results in the blockade of the endosomal TLR signaling pathway in the lymph nodes. (<b>a</b>) The expression of total proteins related to the endosomal TLR signaling was analyzed by Western blotting using lymph nodes from female lupus-prone mice (MRL/<span class="html-italic">lpr</span>). (<b>b</b>) Western blot data were quantified using a densitometer (*<span class="html-italic">P</span> &lt; 0.05, Mann–Whitney <span class="html-italic">U</span> test). n = 4 mice/group. The experiments were repeated at least twice. TIP1, Toll-like receptor inhibitor peptide 1; MyD88, myeloid differentiation factor 88; IRAK4, interleukin-1 receptor-associated kinase 4; TRAF3, tumor necrosis factor receptor-associated factor 3; TRAF6, tumor necrosis factor receptor-associated factor 6.</p>
Full article ">Figure 6
<p>Inhibition of the Toll-like receptor (TLR) signaling pathway in peripheral blood mononuclear cells (PBMCs) of patients with systemic lupus erythematosus (SLE). (<b>a</b>) The expression of the proteins related to the TLR signaling was analyzed by Western blotting using PBMCs from patients with SLE (n = 3) and healthy subjects (HSs; n = 3). All 3 SLE patients were taking hydroxychloroquine 200 mg/d or 300 mg/d. Two SLE patients were not taking corticosteroids except one SLE patient taking prednisolone 2.5 mg/d. (<b>b</b>) Western blot data were quantified using a densitometer (* <span class="html-italic">p</span> &lt; 0.05, Mann–Whitney <span class="html-italic">U</span> test). The experiments were repeated at least twice. TIP1, Toll-like receptor inhibitor peptide 1; MyD88, myeloid differentiation factor 88; IRAK4, interleukin-1 receptor-associated kinase 4; TRAF3, tumor necrosis factor receptor-associated factor 3; TRAF6, tumor necrosis factor receptor-associated factor 6; IRF7, interferon regulatory factor 7; IFN-α, interferon-α.</p>
Full article ">
21 pages, 3475 KiB  
Article
TWEAKing the Hippocampus: The Effects of TWEAK on the Genomic Fabric of the Hippocampus in a Neuropsychiatric Lupus Mouse Model
by Dumitru A. Iacobas, Jing Wen, Sanda Iacobas, Chaim Putterman and Noa Schwartz
Genes 2021, 12(8), 1172; https://doi.org/10.3390/genes12081172 - 29 Jul 2021
Cited by 6 | Viewed by 2468
Abstract
Neuropsychiatric manifestations of systemic lupus erythematosus (SLE), specifically cognitive dysfunction and mood disorders, are widely prevalent in SLE patients, and yet poorly understood. TNF-like weak inducer of apoptosis (TWEAK) has previously been implicated in the pathogenesis of neuropsychiatric lupus (NPSLE), and we have [...] Read more.
Neuropsychiatric manifestations of systemic lupus erythematosus (SLE), specifically cognitive dysfunction and mood disorders, are widely prevalent in SLE patients, and yet poorly understood. TNF-like weak inducer of apoptosis (TWEAK) has previously been implicated in the pathogenesis of neuropsychiatric lupus (NPSLE), and we have recently shown its effects on the transcriptome of the cortex of the lupus-prone mice model MRL/lpr. As the hippocampus is thought to be an important focus of NPSLE processes, we explored the TWEAK-induced transcriptional changes that occur in the hippocampus, and isolated several genes (Dnajc28, Syne2, transthyretin) and pathways (PI3K-AKT, as well as chemokine-signaling and neurotransmission pathways) that are most differentially affected by TWEAK activation. While the functional roles of these genes and pathways within NPSLE need to be further investigated, an interesting link between neuroinflammation and neurodegeneration appears to emerge, which may prove to be a promising novel direction in NPSLE research. Full article
(This article belongs to the Special Issue Genomic Fabric Remodeling in Neurological Disorders)
Show Figures

Figure 1

Figure 1
<p>The independent characteristics of the first 50 alphabetically ordered hippocampal PI3K-AKT genes in the three phenotypes. (<b>a</b>) Average expression level (AVE); (<b>b</b>) Relative expression variability (REV); (<b>c</b>) Pair-wise product-momentum correlation coefficient (COR) of PI3K-AKT genes with <span class="html-italic">Tnfrsf12a.</span> Red/green arrows indicate the statistically significant (<span class="html-italic">p</span> &lt; 0.05) correlations in the MRL/lpr and MRL/+ mice. With AVE = 262 and REV = 193, <span class="html-italic">Akt2</span> is the most remarkable gene of the selection in the hippocampus of the MRL/lpr mice.</p>
Full article ">Figure 2
<p>Measures of differential expression of the first 50 alphabetically ordered hippocampal PI3K-AKT genes in the MRL/lpr mouse with respect to the two controls. (<b>a</b>) Uniform contribution (+1 or −1) of the significantly regulated genes. (<b>b</b>) Fold-change (negative for down-regulation) of all genes within the selection. (<b>c</b>) Weighted Individual (gene) Regulation (WIR). In addition to the net fold-change, WIR considers also the reference expression level of the gene and the statistical confidence (1 − <span class="html-italic">p</span>-value) of its regulation. Taken together, it is a comprehensive measure of the relative contribution of the gene to the model’s transcriptome.</p>
Full article ">Figure 3
<p>Overall transcriptomic differences in the analyzed pathways among the three phenotypes. (<b>a</b>) Percentages of up- and down-regulated genes in selected pathways for indicated comparisons. (<b>b</b>) Weighted Pathway Regulation (WPR) of selected pathways. AKT, PI3K-AKT pathway; CHS, chemokine signaling pathway; CHO, cholinergic neurotransmission; DOP, dopaminergic neurotransmission; GAB, GABAergic neurotransmission, GLU, glutamatergic neurotransmission; SER, serotonergic neurotransmission.</p>
Full article ">Figure 4
<p>Phenotype-dependent transcriptomic network of AKT genes with their KEGG-derived activator and inhibitor genes in the hippocampus. Red/blue lines depict statistically significant (<span class="html-italic">p</span> &lt; 0.05) expression synergism/antagonism between the linked genes. Red line indicates synergism, and blue line antagonism between the 2 genes.</p>
Full article ">Figure 5
<p>Significant in-phase and anti-phase expression of PI3K-AKT genes between the cortex and hippocampus in each phenotype. Red/blue lines indicate the genes that are significantly expressed in-phase and anti-phase between the two regions. Red numbers indicate percentages of in-phase expression, and blue numbers indicate percentages of anti-phase expression.</p>
Full article ">Figure 6
<p>Significant in-phase and anti-phase expression of neurotransmission genes between the cortex and hippocampus in each phenotype. Red/blue lines indicate the genes that are significantly expressed in-phase and antiphase between the two regions. Red numbers indicate percentages of in-phase, and blue numbers of anti-phase expressions.</p>
Full article ">
Back to TopTop