[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
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (233)

Search Parameters:
Keywords = lupus nephritis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 4645 KiB  
Article
Chronic Low-Level IFN-γ Expression Disrupts Mitochondrial Complex I Activity in Renal Macrophages: An Early Mechanistic Driver of Lupus Nephritis Pathogenesis
by Heekyong R. Bae, Su-Kyung Shin, Ji-Yoon Lee, Yeo Jin Ko, Suntae Kim, Howard A. Young and Eun-Young Kwon
Int. J. Mol. Sci. 2025, 26(1), 63; https://doi.org/10.3390/ijms26010063 - 25 Dec 2024
Abstract
Mitochondrial dysfunction and macrophage dysregulation are well recognized as significant contributors to the pathogenesis of autoimmune diseases. However, the detailed mechanisms connecting these two factors remain poorly understood. This study hypothesizes that low but chronic interferon-gamma (IFN-γ) plays a critical role in these [...] Read more.
Mitochondrial dysfunction and macrophage dysregulation are well recognized as significant contributors to the pathogenesis of autoimmune diseases. However, the detailed mechanisms connecting these two factors remain poorly understood. This study hypothesizes that low but chronic interferon-gamma (IFN-γ) plays a critical role in these processes. To explore this, we utilized ARE-Del mice, a model characterized by sustained low-level IFN-γ expression and lupus nephritis (LN)-like symptoms. Age- and tissue-dependent gene expression analyses in ARE-Del mice revealed significant suppression of mitochondrial complex I components and activities, particularly in the kidneys. The genotype-dependent suppression of mitochondrial complex I indicates early disruption, which leads to macrophage dysfunction. Notably, remission restored gene expression of mitochondrial complex I and macrophage dysfunction in isolated renal macrophages from NZB/W lupus-prone mice. These findings suggest that chronic low-level IFN-γ disrupts mitochondrial complex I activity in macrophages, highlighting its role in the early pathogenesis of autoimmune diseases like lupus nephritis. This provides new insights into the molecular interactions underlying autoimmune pathogenesis and suggests potential targets for therapeutic intervention. Full article
(This article belongs to the Special Issue Cytokines in Inflammatory Signaling: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Time- and tissue-dependent hallmark gene set analysis using pre-ranked GSEA in ARE-Del-/- mice over time. (<b>A</b>) Hallmark gene set analysis at 3, 6, and 12 weeks in blood, spleen, thymus, and kidney of ARE-Del-/- mice. The dot size corresponds to the significance level (Sig), represented as −log10(FDR <span class="html-italic">q</span>-value), while the dot color indicates the NES. Blue squares highlight common inflammatory responses. (<b>B</b>) Heatmaps of IFN-γ response genes at 3, 6, and 12 weeks in blood, spleen, thymus, and kidney from ARE-Del-/- mice.</p>
Full article ">Figure 2
<p>Dysregulated mitochondrial complex I activity in the kidney of ARE-Del-/- mice. (<b>A</b>) Tissue-dependent suppression of mitochondrial complex I activity in ARE-Del-/- mice. Dot plots demonstrating the enrichment of gene sets related to mitochondrial respiratory complexes and mitochondrial NADH and ATP responses in blood, spleen, thymus, and kidney of ARE-Del-/- mice at 3, 6, and 12 weeks of age. The dot size corresponds to the significance level (Sig), represented as −log10(FDR <span class="html-italic">q</span>-value), while the dot color indicates the NES. (<b>B</b>) Comparison of mitochondrial complex I activity in the kidneys between ARE-Del-/- vs. ARE-Del+/- mice at 3, 6, 12 weeks of age. Dot plots demonstrating the enrichment of gene sets related to mitochondrial respiratory complex, mitochondrial NADH and ATP, and oxidative stress and abnormal macrophages. The dot size corresponds to the significance level (Sig), represented as −log10(FDR <span class="html-italic">q</span>-value), while the dot color indicates the NES. Columns represent enrichment changes: KOs (ARE-Del-/- vs. control littermates), Hets (ARE-Del+/- vs. control littermates), and KOs vs. Hets (ARE-Del-/- vs. ARE-Del+/- mice).</p>
Full article ">Figure 3
<p>Recovery of inflammatory responses and complex I activity by remission in isolated renal macrophages from NZB/W lupus-prone mice. (<b>A</b>) Dot plots illustrating the changes in hallmark gene sets, comparing the groups of G1_Sick (the active phase of lupus nephritis) vs. G2_Control (during the onset of lupus) and G3_Remission (post-induction of remission) vs. G1_Sick. The y-axis represents fifty hallmark pathways. The count of genes significantly enriched in each pathway, determined by the GSEA algorithm, is shown on the x-axis. Dot size indicates significance through −log10(FDR <span class="html-italic">q</span>-value), while dot color reflects the ES, with red denoting a positive score and blue indicating a negative score. (<b>B</b>) Representative positively enriched plots resulting from GSEA of gene sets associated with IFN-γ, IFN-α, IL-6, and TNF-α responses, comparing the groups of G1 vs. G2 and G3 vs. G1. The baseline (0.0) of the enrichment score is indicated with a red-colored line. The heatmaps illustrate the expression levels of highly enriched genes within each gene set, allowing a comparison between two groups. (<b>C</b>) Dot plots demonstrating the enrichment of gene sets related to mitochondrial respiratory complex, and mitochondrial NADH and ATP, and oxidative stress and abnormal macrophages comparing the groups of G1 vs. G2 and G3 vs. G1. (<b>D</b>) Heatmaps displaying the expression levels of highly enriched genes within each gene set, allowing a comparison between two groups the groups of G1 vs. G2 and G3 vs. G1.</p>
Full article ">
19 pages, 5928 KiB  
Article
The Influence of Anti-ETAR and Anti-CXCR3 Antibody Levels on the Course of Specific Glomerulonephritis Types
by Maciej Szymczak, Harald Heidecke, Marcelina Żabińska, Łucja Janek, Jakub Wronowicz, Krzysztof Kujawa, Karolina Bukowiec-Marek, Tomasz Gołębiowski, Karolina Skalec, Kai Schulze-Forster, Andrzej Konieczny and Mirosław Banasik
J. Clin. Med. 2024, 13(24), 7752; https://doi.org/10.3390/jcm13247752 - 19 Dec 2024
Viewed by 243
Abstract
Background: Anti-ETAR (endothelin A receptor) antibodies and anti-CXCR3 (C-X-C motif chemokine receptor 3) antibodies are types of non-HLA (human leukocyte antigens) antibodies that could have some influence on the course of glomerulonephritis. The authors aimed to study the influence of these antibodies’ levels [...] Read more.
Background: Anti-ETAR (endothelin A receptor) antibodies and anti-CXCR3 (C-X-C motif chemokine receptor 3) antibodies are types of non-HLA (human leukocyte antigens) antibodies that could have some influence on the course of glomerulonephritis. The authors aimed to study the influence of these antibodies’ levels on the course of specific glomerulonephritis types. Methods: We evaluated the anti-ETAR and anti-CXCR3 antibody levels in the serum of patients with membranous nephropathy (n = 18), focal and segmental glomerulosclerosis (FSGS) (n = 25), systemic lupus erythematosus (n = 17), IgA nephropathy (n = 14), mesangiocapillary glomerulonephritis (n = 6), anti-neutrophil cytoplasmic antibodies (c-ANCA) vasculitis (n = 40), and perinuclear anti-neutrophil cytoplasmic antibodies (p-ANCA) vasculitis (n = 16), and we compared their levels with the control group (n = 22). Next, we observed the patients’ clinical parameters (serum creatinine, albumin, total protein) for 2 years and checked the correlation of the clinical course markers with basic receptor antibody level. Results: Our results indicate lower anti-ETAR antibody levels in patients with FSGS and IgA nephropathy compared to the control group. Both types of antibodies correlated with creatinine levels after 2 years of observation in IgA nephropathy. Both types of antibodies seemed to negatively influence the total protein and albumin levels in systemic lupus erythematosus. Conclusions: This prospective observation showed that anti-ETAR and anti-CXCR 3 antibody levels are connected with the course of IgA nephropathy and lupus nephritis. Full article
(This article belongs to the Special Issue Glomerulonephritis: Current Diagnosis, Treatment and Future Options)
Show Figures

Figure 1

Figure 1
<p>Flowchart presenting the process of including in the study.</p>
Full article ">Figure 2
<p>Median ETAR antibody levels in specific glomerular diseases (U/mL). Key: 1—membranous nephropathy; 2—focal and segmental glomerulosclerosis; 3—lupus nephritis; 4—IgA nephropathy; 5—mesangial proliferative (non-IgA) glomerulonephritis; 6—control group; 7—c-ANCA vasculitis; 8—p-ANCA vasculitis. The groups with values in red were statistically lower than the control group.</p>
Full article ">Figure 3
<p>Median CXCR 3 antibodies in specific glomerular diseases (U/mL). Key: 1—membranous nephropathy; 2—focal and segmental glomerulosclerosis; 3—lupus nephritis; 4—IgA nephropathy; 5—mesangial proliferative (non-IgA) glomerulonephritis; 6—control group; 7—c-ANCA vasculitis; 8—p-ANCA vasculitis.</p>
Full article ">Figure 4
<p>Correlation between basic serum anti-ETAR antibody level and serum creatinine after 2 years of observation in membranous nephropathy patients (r = 0.55; <span class="html-italic">p</span> = 0.03). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 5
<p>Correlation between basic serum anti-ETAR antibody level and serum creatinine after 2 years of observation in IgA nephropathy patients (r = 0.57; <span class="html-italic">p</span> = 0.03). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 6
<p>Correlation between basic serum anti-ETAR antibody level and serum total protein level after 3 months of observation in lupus nephritis group (r = −0.54; <span class="html-italic">p</span> = 0.03). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 7
<p>Correlation between basic serum anti-ETAR antibody level and serum albumin level after 3 months of observation in lupus nephritis group (r = −0.62; <span class="html-italic">p</span> = 0.01). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 8
<p>Correlation between basic serum anti-ETAR antibody level and serum albumin level after 6 months of observation in lupus nephritis group (r = −0.57; <span class="html-italic">p</span> = 0.03). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 9
<p>Correlation between basic serum anti-CXCR3 antibody level and basic serum total protein level in focal and segmental glomerulosclerosis group (r = 0.50; <span class="html-italic">p</span> = 0.009). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 10
<p>Correlation between basic serum anti-CXCR3 antibody level and serum total protein level after 1 month of observation in focal and segmental glomerulosclerosis group (r = 0.49; <span class="html-italic">p</span> = 0.02). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 11
<p>Correlation between basic serum anti-CXCR3 antibody level and serum total protein level after 3 months of observation in focal and segmental glomerulosclerosis group (r = 0.43; <span class="html-italic">p</span> = 0.04). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 12
<p>Correlation between basic serum anti-CXCR3 antibody level and serum creatinine level after 2 years of observation in IgA nephropathy group (r = 0.65; <span class="html-italic">p</span> = 0.01). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 13
<p>Correlation between basic serum anti-CXCR3 antibody level and serum total protein level after 2 years of observation in lupus nephritis group (r = −0.58; <span class="html-italic">p</span> = 0.03). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 14
<p>Correlation between basic serum anti-CXCR3 antibody level and basic serum albumin level in lupus nephritis group (r = −0.53; <span class="html-italic">p</span> = 0.02). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 15
<p>Correlation between basic serum anti-CXCR3 antibody level and albumin level after 1 month of observation in lupus nephritis group (r = −0.51; <span class="html-italic">p</span> = 0.04). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 16
<p>Correlation between basic serum ETAR antibody level and serum CXCR3 antibody level in lupus nephritis group (r = 0.58; <span class="html-italic">p</span> = 0.01). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">Figure 17
<p>Correlation between basic serum ETAR antibody level and serum CXCR3 antibody level in IgA nephropathy group (r = 0.76; <span class="html-italic">p</span> = 0.001). Blue dots represent particular patients. Blue dotted line—trend line.</p>
Full article ">
11 pages, 507 KiB  
Perspective
Antibodies Against Anti-Oxidant Enzymes in Autoimmune Glomerulonephritis and in Antibody-Mediated Graft Rejection
by Maurizio Bruschi, Giovanni Candiano, Andrea Petretto, Andrea Angeletti, Pier Luigi Meroni, Marco Prunotto and Gian Marco Ghiggeri
Antioxidants 2024, 13(12), 1519; https://doi.org/10.3390/antiox13121519 - 12 Dec 2024
Viewed by 384
Abstract
Historically, oxidants have been considered mechanisms of glomerulonephritis, but a direct cause–effect correlation has never been demonstrated. Several findings in the experimental model of autoimmune conditions with renal manifestations point to the up-regulation of an oxidant/anti-oxidant system after the initial deposition of autoantibodies [...] Read more.
Historically, oxidants have been considered mechanisms of glomerulonephritis, but a direct cause–effect correlation has never been demonstrated. Several findings in the experimental model of autoimmune conditions with renal manifestations point to the up-regulation of an oxidant/anti-oxidant system after the initial deposition of autoantibodies in glomeruli. Traces of oxidants in glomeruli cannot be directly measured for their rapid metabolism, while indirect proof of their implications is derived from the observation that Superoxide Oxidase 2 (SOD2) is generated by podocytes after autoimmune stress. The up-regulation of other anti-oxidant systems takes place as well. Here, we discuss the concept that a second wave of antibodies targeting SOD2 is generated in autoimmune glomerulonephritis and may negatively influence the clinical outcome. Circulating and renal deposits of anti-SOD2 antibodies have been detected in patients with membranous nephropathy and lupus nephritis, two main examples of autoimmune disease of the kidney, which correlate with the clinical outcome. The presence of anti-SOD2 antibodies in circulation and in the kidney has been interpreted as a mechanism which modifies the normal tissue response to oxidative stress. Overall, these findings repropose the role of the oxidant/anti-oxidant balance in autoimmune glomerulonephritis. The same conclusion on the oxidant/anti-oxidant balance may be proposed in renal transplant. Patients receiving a renal graft may develop antibodies specific for Glutathione Synthetase (GST), which modulates the amount of GST disposable for rapid scavenging of reactive oxygen species (ROS). The presence of anti-GST antibodies in serum is a major cause of rejection. The perspective is to utilize molecules with known anti-oxidant effects to modulate the anti-oxidative response in autoimmune pathology of the kidney. A lot of molecules with known anti-oxidant effects can be utilized, many of which have already been proven effective in animal models of autoimmune glomerulonephritis. Many molecules with anti-oxidant activity are natural products; in some cases, they are constituents of diets. Owing to the simplicity of these drugs and the absence of important adverse effects, many anti-oxidants could be directly utilized in human beings. Full article
(This article belongs to the Special Issue Oxidative Stress in Renal Health)
Show Figures

Figure 1

Figure 1
<p>Patients with MN (285 overall) were studied at diagnosis and during a long follow-up of 46 months with multiple clinical approaches. At diagnosis, 182 (64%), 8 (3%), and 95 (33%) patients were anti-PLA2R1+, anti-THSD7A+ and double-negative, respectively. Kaplan–Meier analysis showed that anti-PLA2R1 and anti-SOD2 antibodies at diagnosis were each independently associated with poor clinical outcomes. Combined positivity for anti-PLA2R1 and anti-SOD conferred maximal risk.</p>
Full article ">
14 pages, 1522 KiB  
Article
Urinary Immune Complexes Reflect Renal Pathology in Lupus Nephritis
by Chenling Tang, Aygun Teymur and Tianfu Wu
Diagnostics 2024, 14(24), 2787; https://doi.org/10.3390/diagnostics14242787 - 12 Dec 2024
Viewed by 377
Abstract
Background/Objectives: Lupus nephritis (LN) is a serious complication of systemic lupus erythematosus (SLE), involving immune complex deposition in the kidneys. While renal biopsy is the diagnostic gold standard, its invasiveness limits frequent use, driving the need for non-invasive urinary biomarkers to monitor disease [...] Read more.
Background/Objectives: Lupus nephritis (LN) is a serious complication of systemic lupus erythematosus (SLE), involving immune complex deposition in the kidneys. While renal biopsy is the diagnostic gold standard, its invasiveness limits frequent use, driving the need for non-invasive urinary biomarkers to monitor disease progression and response to treatment. This study aimed to identify and validate urinary biomarkers for LN. Methods: Data from 10 LN-related omics databases, including urine, PBMCs, and kidney tissue, were analyzed. Differentially expressed proteins (DEPs) and genes (DEGs) were identified, and candidate biomarkers were validated via ELISA in an independent cohort of 87 urine samples. Results: We identified 78 biomarkers, with 14 overlapping across transcriptomic categories. Novel urinary biomarkers, including SERPING1, SLPI, and CD48, were validated. Urinary CD163, VCAM1, and ALCAM levels showed significant differences between LN and healthy controls, while urinary immune complexes (ICx) demonstrated superior diagnostic performance, with urinary ALCAM-ICx and CCL21-ICx achieving the highest AUC values. Conclusions: Our findings highlight the potential of urinary immune complexes and antigens as non-invasive biomarkers for LN. ALCAM, CD163, and SERPING1-ICx, in particular, were found as promising candidates for a urinary biomarker panel to aid in the diagnosis and monitoring of LN. Full article
(This article belongs to the Section Clinical Laboratory Medicine)
Show Figures

Figure 1

Figure 1
<p>Omics Identification of LN concurrence biomarkers. (<b>A</b>) Venn diagram showing overlap of differentially expressed proteins (DEPs) and genes (DEGs) from urine protein array, kidney scRNA, kidney RNA-array, PBMC scRNA, and PBMC RNA-array datasets. (<b>B</b>) Heatmap displaying the expression levels (log2 fold change) of selected biomarkers, colored based on their frequency of occurrence across the databases. (<b>C</b>) Bar graph depicting the number of biomarkers with co-occurrence in 1 to 4 datasets. (<b>D</b>) Protein–protein interaction network of top DEPs/DEGs identified, visualized using STRING analysis.</p>
Full article ">Figure 2
<p>ELISA screening of potential urine biomarker candidates of LN. Box plots showing the results of ELISA validation for selected urinary antigens (top row) and their corresponding immune complexes (ICx, middle and bottom rows) across lupus nephritis (LN), chronic kidney disease (CKD), and healthy control (HC) groups. Biomarkers analyzed include CD163, SPP1, NGAL, ALCAM, VCAM1, SERPING1, BAFF, CCL21, TFPI, VSIG4, SLPI, and uCreatinine. All urinary biomarker levels (shown in Y-axis) were normalized by urinary creatinine. Statistical significance between groups is indicated by asterisks (* <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; **** <span class="html-italic">p</span> &lt; 0.00001).</p>
Full article ">Figure 3
<p>Diagnostic potential of urinary antigens and ICx. (<b>A</b>) Bar graphs displaying the area under the curve (AUC) values for individual biomarkers and immune complexes (ICx) in distinguishing LN from healthy controls (HC), LN flare from remission, LN from CKD, and CKD from HC. (<b>B</b>) Comparison of six machine-learning models for classifying LN versus HC, showing the average AUC values from 3-fold, 10-repeats. Models include random forest, support vector machine, K-nearest neighbors, decision tree, linear discriminant analysis, and logistic regression. (<b>C</b>) Heatmap illustrating the Spearman correlation between urinary biomarkers and clinical/pathological indices, including SLEDAI, AI (activity index), CI (chronicity index), age, WBC, uPCR, serum creatinine, and uRBC. Statistical significance is denoted by asterisks (* <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; **** <span class="html-italic">p</span> &lt; 0.00001).</p>
Full article ">
16 pages, 1528 KiB  
Article
Circulating B Lymphocyte Subsets in Patients with Systemic Lupus Erythematosus
by Joanna Kosałka-Węgiel, Bogdan Jakieła, Radosław Dziedzic, Mamert Milewski, Andżelika Siwiec-Koźlik, Lech Zaręba, Stanisława Bazan-Socha, Marek Sanak, Jacek Musiał and Mariusz Korkosz
Medicina 2024, 60(12), 1994; https://doi.org/10.3390/medicina60121994 - 2 Dec 2024
Viewed by 622
Abstract
Background/Objectives: Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the abnormal activation of autoreactive T and B cells, autoantibody production, complement activation, and immune-complex deposition, resulting in tissue damage. However, data on immunologic disturbances in SLE, particularly regarding flares, are [...] Read more.
Background/Objectives: Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the abnormal activation of autoreactive T and B cells, autoantibody production, complement activation, and immune-complex deposition, resulting in tissue damage. However, data on immunologic disturbances in SLE, particularly regarding flares, are scarce. Methods: We investigated 35 patients with SLE: 12 (34.3%) with disease exacerbation (SLE disease activity index [SLEDAI] ≥ 5 points) and 23 (65.7%) in remission (SLEDAI < 5 points). All patients met the 2019 EULAR/ACR SLE criteria. Flow cytometry was used to identify B cell subsets, including memory B cells. Results: In the whole patient group, SLEDAI was positively related to the percentage of transitional/regulatory B cells (r = 0.38, p = 0.034). Some lymphocyte subsets correlated with complement levels, e.g., the percentage of naïve and memory B cells showed associations with C3c complement (r = 0.43, p = 0.018 and r = −0.45, p = 0.016, respectively). Furthermore, regarding inflammatory markers, we found associations between C-reactive protein and the percentage of plasmablasts (r = 0.40, p = 0.026) and plasmocytes (r = 0.44, p = 0.017). Finally, the percentage of plasmablasts correlated with SLE duration (r = 0.42, p = 0.016). In the follow-up analysis, during a median observation of 5 years, 5 out of the initially 23 inactive SLE patients developed a disease flare. They were characterized by longer disease duration stated in the beginning compared to patients who remained in remission (p = 0.019). Conclusions: Our study highlights significant associations between various B cell subsets and SLE disease activity. A more personalized approach to indicate patients with SLE at a higher risk of lupus flares is crucial for better management. Full article
(This article belongs to the Special Issue Recent Advances in Autoimmune Rheumatic Diseases: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Flow cytometry staining of B cell subsets. (<b>a</b>) Schematic diagram showing the flow cytometry (FC) gating strategy used to identify major B cell subsets. Samples of peripheral blood mononuclear cells (PMBCs) were stained with pre-mixed antibodies (CD19-PerCP/Cy5.5, CD38-APC, CD27-PE, CD24-PE/Cy7, IgD-V450, CD21-FITC), and analyzed in FC (&gt;10<sup>3</sup> events in B cell gate recorded). (<b>b</b>,<b>c</b>) Representative dot plots showing gates and analysis flowchart. (<b>b</b>) CD19+ singlets (R0 + R1, only FSC-A/W gate shown) were first analyzed to distinguish the CD38++ CD27++ subset of plasma cells and plasmablasts (R4 region). (<b>c</b>) The remaining non-plasma cells were gated (R3) to identify naïve B- B cells, non-switched memory (NSM) B cells, and switched-memory (SM) B cells. Transitional B cells (TrB)/regulatory B cells (Breg) were identified as CD38++ CD24++ using an oblique gate (R8). TrB cells showed the following phenotype: CD19+ CD38++ CD24++ CD27− IgD(low/+) CD21(low/+). (<b>d</b>) An additional sample (stained with CD19-PerCP/Cy5.5, CD38-FITC, CD27-PE, CD138-PE/Cy7, IgD-V450, and IgM-APC) was used to distinguish between plasmablasts (CD138−) and plasma cells (CD138+). These two subsets were CD27++ and were negative for membrane IgD and IgM.</p>
Full article ">
9 pages, 5649 KiB  
Case Report
Delayed Diagnosis of Disseminated Invasive Aspergillosis with Purulent Myocarditis in an Immunocompromised Host
by Mark Londema, Maarten W. N. Nijsten, Joost Bart, Janke S. Wiegersma, Bhanu N. M. Sinha and Douwe F. Postma
Infect. Dis. Rep. 2024, 16(6), 1182-1190; https://doi.org/10.3390/idr16060093 - 30 Nov 2024
Viewed by 471
Abstract
Introduction: Invasive aspergillosis (IA) is an opportunistic fungal infection that typically occurs in the immunocompromised host and is associated with severe morbidity and mortality. Myocardial abscess formation is seldomly described. Detailed Case Description: We present a case of IA with purulent myocarditis. The [...] Read more.
Introduction: Invasive aspergillosis (IA) is an opportunistic fungal infection that typically occurs in the immunocompromised host and is associated with severe morbidity and mortality. Myocardial abscess formation is seldomly described. Detailed Case Description: We present a case of IA with purulent myocarditis. The patient was on long-term high-dose corticosteroid and mycophenolate mofetil therapy for severe lupus nephritis. After multiple visits to his general practitioner and nephrologist for general malaise, he was admitted to our hospital with visual complaints. Within several days, he developed atrial fibrillation, respiratory insufficiency, and, finally, a decreased level of consciousness. After admission to the intensive care unit, the broncho alveolar lavage (BAL) fluid galactomannan (GM) index was normal, but the serum GM index was severely elevated. Despite initiation of antifungal therapy, the patient passed away shortly thereafter. Autopsy revealed massive intracranial hemorrhage and disseminated IA affecting the lungs, brain, and myocardium, with macroscopic myocardial abscess formation. Discussion: This classic case of diagnostic uncertainty illustrates how invasive fungal infections can progress to disseminated disease while showing nonspecific symptoms only. It emphasizes the importance of vigilance for opportunistic fungal infections in a growing category of immunocompromised patients. Conclusion: Clinicians should have a low threshold of suspicion for fungal infections in patients on combination immunosuppressive medication, such as high-dose corticosteroid therapy in combination with T-cell inhibitors like MMF. Full article
(This article belongs to the Section Fungal Infections)
Show Figures

Figure 1

Figure 1
<p>Transverse section through both ventricles, showing multiple myocardial Aspergillus abscesses.</p>
Full article ">Figure 2
<p>Hematoxylin and eosin staining. Black bar represents 500 μm. Micrograph of the heart, demonstrating heavy intramyocardial inflammation with abscess formation, caused by Aspergillus fumigatus. a: myocardium; b: epicardial fatty tissue; c: lumen of a branch of the coronary arterial system; d: inflammatory infiltrate and abscess wall; e: abscess cavity; rectangle: depicts area that is presented in <a href="#idr-16-00093-f003" class="html-fig">Figure 3</a>.</p>
Full article ">Figure 3
<p>Grocott silver staining. Black bar represents 50 μm. Detailed image of the abscess wall, demonstrating the branching Aspergillus hyphae.</p>
Full article ">Figure 4
<p>Hematoxylin and eosin staining. Black bar represents 2 mm. Micrograph of the lung demonstrating abscess with Aspergillus fumigatus. a: (alveolar) lung parenchyma; b: artery; c: vein; d: inflammatory infiltrate; e: abscess also depicted in Grocott stain in <a href="#idr-16-00093-f005" class="html-fig">Figure 5</a>.</p>
Full article ">Figure 5
<p>Grocott silver staining. Black bar represents 50 μm. Detailed image of the lung abscess with abundant branching Aspergillus hyphae.</p>
Full article ">
16 pages, 864 KiB  
Article
Analysis of the Sensitivity and Specificity of Histopathological Findings for Diagnosing Lupus Nephritis
by Epitácio Rafael da Luz Neto, Maria Brandão Tavares, Ana Gabriela de Jesus Torres de Melo, Washington L. C. dos-Santos, Denise Maria Avancini Costa Malheiros and Luís Yu
Diagnostics 2024, 14(23), 2681; https://doi.org/10.3390/diagnostics14232681 - 27 Nov 2024
Viewed by 460
Abstract
Background: Since the introduction of the SLICC criteria in 2012, biopsy-proven lupus nephritis (LN) has been the only independent diagnostic criterion for systemic lupus erythematosus (SLE). This was reaffirmed by the EULAR/ACR in 2019, emphasizing the importance of renal biopsy in LN. However, [...] Read more.
Background: Since the introduction of the SLICC criteria in 2012, biopsy-proven lupus nephritis (LN) has been the only independent diagnostic criterion for systemic lupus erythematosus (SLE). This was reaffirmed by the EULAR/ACR in 2019, emphasizing the importance of renal biopsy in LN. However, the current classification lacks specific histopathological criteria for defining LN. This study describes the histological findings of patients with LN, compares them with those of other glomerular diseases, and evaluates their diagnostic accuracy in a large Latin American population. Methods: This retrospective cohort included 731 kidney biopsies from two distinct academic centers. The patients were divided into two groups as follows: a LN group and a control group comprising patients with membranous nephropathy, IgA nephropathy, membranoproliferative glomerulonephritis, pauci-immune glomerulonephritis, and proliferative glomerulonephritis. Sensitivity and specificity analyses were conducted for various histopathological features. Results: We identified the following five features strongly correlated with LN: mesangial proliferation, subendothelial deposits, C1q staining ≥1+, dominant IgG, and ≥4 positive immunofluorescence elements. Combined, these features yielded an area under the ROC curve of 0.94 (95% CI: 0.91–0.95). These results were validated in a diverse population. In membranous nephropathy, histological features such as mesangial deposits, C1q positivity, and ≥4 positive immunofluorescence elements effectively distinguished class V LN from non-lupus membranous nephropathy, with an area under the ROC curve of 0.85 (95% CI: 0.76–0.93). Conclusions: The combination of mesangial proliferation, subendothelial deposits, C1q staining ≥1+, dominant IgG, and ≥4 positive immunofluorescence elements offer good accuracy for diagnosing renal involvement in SLE in a large Latin American population. In the absence of pathognomonic features, combined criteria are valuable diagnostic tools, particularly when other SLE criteria are lacking. Full article
(This article belongs to the Special Issue Current Issues on Kidney Diseases Diagnosis and Management 2025)
Show Figures

Figure 1

Figure 1
<p>ROC curve to distinguish lupus nephritis and non-lupus glomerulopathies—HCFMUSP. The diagonal line defines an area under the curve of 50%.</p>
Full article ">Figure 2
<p>ROC curve to distinguish lupus nephritis and non-lupus glomerulopathies—HAN. The diagonal line defines an area under the curve of 50%.</p>
Full article ">
15 pages, 1266 KiB  
Review
Technology Innovation for Discovering Renal Autoantibodies in Autoimmune Conditions
by Maurizio Bruschi, Giovanni Candiano, Andrea Petretto, Andrea Angeletti, Pier Luigi Meroni, Marco Prunotto and Gian Marco Ghiggeri
Int. J. Mol. Sci. 2024, 25(23), 12659; https://doi.org/10.3390/ijms252312659 - 25 Nov 2024
Viewed by 563
Abstract
Autoimmune glomerulonephritis is a homogeneous area of renal pathology with clinical relevance in terms of its numerical impact and difficulties in its treatment. Systemic lupus erythematosus/lupus nephritis and membranous nephropathy are the two most frequent autoimmune conditions with clinical relevance. They are characterized [...] Read more.
Autoimmune glomerulonephritis is a homogeneous area of renal pathology with clinical relevance in terms of its numerical impact and difficulties in its treatment. Systemic lupus erythematosus/lupus nephritis and membranous nephropathy are the two most frequent autoimmune conditions with clinical relevance. They are characterized by glomerular deposition of circulating autoantibodies that recognize glomerular antigens. Technologies for studying renal tissue and circulating antibodies have evolved over the years and have culminated with the direct analysis of antigen–antibody complexes in renal bioptic fragments. Initial studies utilized renal microdissection to obtain glomerular tissue. Obtaining immunoprecipitates after partial proteolysis of renal tissue is a recent evolution that eliminates the need for tissue microdissection. New technologies based on ‘super-resolution microscopy’ have added the possibility of a direct analysis of the interaction between circulating autoantibodies and their target antigens in glomeruli. Peptide and protein arrays represent the new frontier for identifying new autoantibodies in circulation. Peptide arrays consist of 7.5 million aligned peptides with 16 amino acids each, which cover the whole human proteome; protein arrays utilize, instead, a chip containing structured proteins, with 26.000 overall. An example of the application of the peptide array is the discovery in membranous nephropathy of many new circulating autoantibodies including formin-like-1, a protein of podosomes that is implicated in macrophage movements. Studies that utilize protein arrays are now in progress and will soon be published. The contribution of new technologies is expected to be relevant for extending our knowledge of the mechanisms involved in the pathogenesis of several autoimmune conditions. They may also add significant tools in clinical settings and modify the therapeutic handling of conditions that are not considered to be autoimmune. Full article
Show Figures

Figure 1

Figure 1
<p>Workflow utilized for characterizing glomerular antibodies microeluted from the kidney. Glomerular microdissection is the first step: (<b>a</b>,<b>b</b>) show a renal bioptic sample before and after microdissection, and (<b>c</b>) shows the glomerulus derived from the procedure. Glomerular extracts are then incubated with podocyte proteins previously separated by 2D electrophoresis and transferred to nitrocellulose membranes. Those spots that are recognized by immunoglobulin glomerular extracts undergo characterization by mass spectrometry.</p>
Full article ">Figure 2
<p>Validation of antibodies microeluted from glomeruli and characterized by immunoblot and mass spectrometry is carried out by immunofluorescence on kidney biopsies. The example presented in this figure is the validation of alpha-enolase as an antigen in patients with lupus nephritis. In this case, alpha-enolase, stained in red, and IgG2, stained in green, have an intense yellow merge, which indicates that the two proteins interact in the tissue. Magnification: ×400.</p>
Full article ">Figure 3
<p>Whole proteome peptide arrays consist of 7,499,126 peptides with 16 amino acids each that together cover the amino acid sequence of all the proteins coded by the human genome. Sera are incubated with all 7,499,126 peptides of the customized array, and the intensity of the relative fluorescence deriving from their interaction is aligned in sequence by informatic technologies to obtain the identification of a unique linear epitope corresponding to a specific protein. This figure shows the application of the peptide arrays for discovering new circulating antibodies in patients with membranous nephropathy, which is a unique example of the application of the array in human pathology [<a href="#B17-ijms-25-12659" class="html-bibr">17</a>].</p>
Full article ">
14 pages, 2537 KiB  
Article
Leflunomide-Induced Weight Loss: Involvement of DAHPS Activity and Synthesis of Aromatic Amino Acids
by Xiaoyu Guo, Kai Wang, Hongli Chen, Na Wang, Dongmei Qiu, Haiyun Huang, Jiyu Luo, Ao Xu, Lingyun Xu, Zejun Yu, Yuanyuan Li and Hongling Zhang
Metabolites 2024, 14(11), 645; https://doi.org/10.3390/metabo14110645 - 20 Nov 2024
Viewed by 782
Abstract
Background/Objectives: Leflunomide, an isoxazole immunosuppressant, is widely used in the treatment of diseases such as rheumatoid arthritis (RA) and psoriatic arthritis (PsA) as well as lupus nephritis (LN). In recent years, clinical data have shown that some patients have obvious weight loss, liver [...] Read more.
Background/Objectives: Leflunomide, an isoxazole immunosuppressant, is widely used in the treatment of diseases such as rheumatoid arthritis (RA) and psoriatic arthritis (PsA) as well as lupus nephritis (LN). In recent years, clinical data have shown that some patients have obvious weight loss, liver injury, and other serious adverse reactions after taking leflunomide. However, the causes and mechanisms by which leflunomide reduces weight are unclear. Methods: Therefore, we used a mouse animal model to administer leflunomide, and we observed that the weight of mice in the leflunomide experimental group was significantly reduced (p < 0.01). In this animal experiment, a metabolomic method was used to analyze the livers of the mice in the experimental group and found that the main difference in terms of metabolic pathways was in the metabolism of aromatic amino acids, and it was confirmed that leflunomide can inhibit the limitations of phenylalanine, tyrosine, and tryptophan biosynthesis. Results: Our study revealed that leflunomide inhibited the activity of DAHPS in the gut microbiota, disrupting the metabolism of phenylalanine, tyrosine, and tryptophan, as well as the metabolism of carbohydrates and lipids. Leflunomide also increased endoplasmic reticulum stress by activating the PERK pathway, thereby promoting CHOP expression and increasing apoptosis-induced liver damage. Conclusions: These effects may be related to the observed weight loss induced by leflunomide. Full article
(This article belongs to the Special Issue The Interplay Between Inflammation and Metabolism in Disease)
Show Figures

Figure 1

Figure 1
<p>The body weight of mice in the leflunomide treatment group decreased significantly. (<b>A</b>) The average daily body weight change curve of the four groups of mice. (<b>B</b>) The average daily food intake of the four groups of mice. (<b>C</b>) Average 11-day food intake of each group (16 mice) of the four groups. (<b>D</b>) Liver weight changes of mice in the four groups after 12 days of drug treatment. (<b>E</b>) Four groups of mice were weighed and sacrificed 12 days after administration to obtain fat for calculating body fat ratio data. *, <span class="html-italic">p</span> &lt; 0.05 and **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Morphological investigations of the liver of mice. (<b>A</b>–<b>C</b>) Scale bar of liver slices from mice in the 30 mg/kg/d group and control group. (<b>A</b>) HE staining of hepatic cells in each group. The control group showed normal morphological manifestation of hepatic cells. The arrangement and size of cells were normal. No steatosis or edema was found. The hepatic cells in the 30 mg/d group exhibited mild changes. The arrangement of cells was chaotic. Slight fat vacuoles could be observed in this group. (<b>B</b>) shows the quantitative data of PAS (periodic acid–Schiff) staining. (<b>C</b>) shows the quantitative data of oil red staining and triglyceride staining. (<b>D</b>,<b>E</b>) The staining results were quantified with Image J to show the levels of triglycerides and polysaccharides in each group. Leflunomide interferes with the synthesis of polysaccharides and triglycerides in the livers of mice. Data are expressed as mean ± SD, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Leflunomide inhibits the biosynthetic pathway of aromatic amino acids in mice. (<b>A</b>) Heat map of the hierarchical clustering analysis of the treatment group to the control group. In the figure, the abscissa represents different experimental groupings, the ordinate represents the metabolic differences in this group, and the color patches at different positions represent the different expression levels of the metabolites at the corresponding positions. (<b>B</b>) Path analysis chart of treatment group to control group. The results of the metabolic pathway analysis are displayed in a bubble chart. Each bubble in the bubble chart represents a metabolic pathway. The abscissa and the size of the bubble indicate the size of the influence factor of the pathway in the topological analysis. The larger the bubble, the greater the influence factor. The vertical axis of the bubble and the color of the bubble indicate the <span class="html-italic">p</span> value of enrichment analysis (negative natural logarithm, namely, In <span class="html-italic">p</span> value).</p>
Full article ">Figure 4
<p>Leflunomide has proliferative toxicity to normal human hepatocytes. (<b>A</b>) Normal human hepatocytes were exposed to leflunomide at 20 µΜ, 50 µΜ, and 100 µΜ concentrations or DMSO, or were left untreated (untreated group: UT) for 12 h. Data are expressed as mean ± SD (n = 3). The concentration of 50% activity against liver cells (IC50 = 203 ± 6.78μM) was obtained from the metered response curve of leflunomide to normal human liver cells. (<b>B</b>) Normal human hepatocytes at a concentration of 10,000/mL were exposed to leflunomide at 20 µΜ for 9 days, and the fluid was changed every 3 days. (<b>C</b>) A total of 1000 normal human hepatocytes were exposed to leflunomide at a concentration of 20 µΜ, clone formation was observed after 9 days, and the medium supplemented with the drug was changed every 3 days. (<b>D</b>) Compared with the control group, the value-added activity of hepatocytes was significantly reduced in the leflunomide group, and this result has statistical significance. Data are expressed as mean ± SD, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Leflunomide can induce endoplasmic reticulum stress. (<b>A</b>) After normal human hepatocytes were exposed to leflunomide at 20 µΜ, 50 µΜ, and 100 µΜ concentrations for 12 h. Immunoblotting experiments showed protein expression levels in cells or tissues and quantified the protein. On the right, there are four groups of mice in the control group and 30 mg/kg/d group randomly selected after 12 days to detect the protein levels in human normal hepatocytes in vitro of the liver. On the left side of the figure, there is the concentration of 20 µM. (<b>B</b>) Liver slices were immunostained with Ki67 and caspase-3 after 12 days in the control group and the 30 mg/kg/d group. The staining was then scored and quantified. Scale bars: left, 50 µm; right, 15 µm. Data are expressed as mean ± SD, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Leflunomide inhibits DAHPS activity in the intestinal flora of mice. (<b>A</b>) The feces of mice collected 12 days after the administration of the test were tested for DAHPS activity in the fecal intestinal flora. (<b>B</b>) Mice were dissected after 12 days of continuous administration, and the contents of the small intestines of mice were removed to detect DAHPS activity in the intestinal flora. Compared with the control group, these results have statistical significance. Data are expressed as mean ± SD, * <span class="html-italic">p</span> &lt;0.05.</p>
Full article ">
16 pages, 11712 KiB  
Article
Oxymatrine Ameliorates Lupus Nephritis by Targeting the YY1-Mediated IL-6/STAT3 Axis
by Haoxing Yuan, Zheng Peng, Honglian Li, Yuzhen Rao, Kunyu Lu, Chan Yang, Chen Cheng and Shuwen Liu
Int. J. Mol. Sci. 2024, 25(22), 12260; https://doi.org/10.3390/ijms252212260 - 14 Nov 2024
Viewed by 852
Abstract
Lupus nephritis (LN) is a severe form of systemic lupus erythematosus (SLE), characterized by inflammation in the renal glomeruli and tubules. Previous research has demonstrated that dihydroartemisinin (DHA) can reduce inflammatory damage in LN mouse models. Oxymatrine, which has similar biological properties to [...] Read more.
Lupus nephritis (LN) is a severe form of systemic lupus erythematosus (SLE), characterized by inflammation in the renal glomeruli and tubules. Previous research has demonstrated that dihydroartemisinin (DHA) can reduce inflammatory damage in LN mouse models. Oxymatrine, which has similar biological properties to DHA, may also provide therapeutic benefits. This study aims to investigate the effects of oxymatrine on LN using a murine model and examines its molecular mechanisms through an analysis of microarray datasets from LN patients. The analysis identified differentially expressed genes (DEGs) in renal tissues, regulated by the transcription factor Yin Yang 1 (YY1), which was found to be significantly upregulated in LN patient kidneys. The results indicate that oxymatrine targets the YY1/IL-6/STAT3 signaling pathway. In cell models simulating renal inflammation, oxymatrine reduced YY1 expression and inhibited the secretion of inflammatory factors (IFs), thereby diminishing inflammation. YY1 is crucial in modulating IFs’ secretion and contributing to LN pathogenesis. Additionally, oxymatrine’s interaction with YY1, leading to its downregulation, appears to be a key mechanism in alleviating LN symptoms. These findings support oxymatrine as a promising therapeutic agent for LN, offering new avenues for treating this autoimmune kidney disorder. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Oxymatrine alleviates splenomegaly, proteinuria, and inflammation in mice with lupus nephritis (LN). (<b>A</b>) Schematic representation of the mouse study design. (<b>B</b>) Representative images of spleens from the mice. (<b>C</b>) Weight of the mice. (<b>D</b>) Spleen weight index (total spleen weight/body weight) of the mice. (<b>E</b>) 24 h proteinuria levels in the mice. (<b>F</b>) Proportion of T lymphocyte cells. (<b>G</b>) Levels of dsDNA in the serum of the mice. (<b>H</b>) Levels of IL-6, IL-1β, and TNF-α in the serum of the mice. Each bar represents the mean ± SEM. ns, no significance, * <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; one-way ANOVA was used to compare multiple groups.</p>
Full article ">Figure 2
<p>Oxymatrine improved the nephritis in mice with LN. (<b>A</b>) Serum levels of S-cr in mice. (<b>B</b>) Serum levels of BUN in mice. (<b>C</b>) Hematoxylin and eosin staining of kidney sections from the indicated groups. Scale bar = 20 μm. (<b>D</b>) Periodic Acid-Schiff staining of kidney sections from the indicated groups. Scale bar = 20 μm. (<b>E</b>) Immunoglobulin G (IgG) staining of kidney sections from the indicated groups. Scale bar = 10 μm. (<b>F</b>) Component 3 (C3) staining of kidney sections from the indicated groups. Scale bar = 10 μm. (<b>G</b>) Staining of IL-6, IL-1β, and TNF-α in kidney sections from the indicated groups. Scale bar = 20 μm. Black arrowheads indicate immune complexes, and red boxes indicate C3. Each bar represents the mean ± SEM. ns, no significance, * <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; one-way ANOVA was used to compare multiple groups.</p>
Full article ">Figure 3
<p>Oxymatrine modulates the YY1/IL-6/STAT3 pathway to alleviate inflammation in LN. (<b>A</b>) Volcano plots displaying differentially expressed genes (DEGs) in LN versus control (<span class="html-italic">n</span> = 28,439). (<b>B</b>) TRRUST analysis of DEGs from panel A. (<b>C</b>) GSEA of IL-6 Jak STAT3 signaling pathway. FDR, false discovery rate; NES, normalized enrichment score. (<b>D</b>) Relative protein levels of Yin Yang 1 (YY1) in the kidneys of LN patients assessed by IHC. Scale bar = 20 μm. (<b>E</b>,<b>F</b>) Western blot analysis of YY1 levels in the kidneys of wild-type mice and from MRL/lpr mice. The signal densities of YY1 were normalized to that of β-actin. (<b>G</b>) Quantitative real-time PCR (qRT–PCR) analysis of YY1 mRNA levels in the kidneys of wild-type mice and from MRL/lpr mice. (<b>H</b>) Western blot analysis of YY1 levels and phosphorylation levels of Stat3 in the kidneys of mice. The signal densities of YY1 were normalized to that of β-actin. Each bar represents the mean ± SEM. * <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; <span class="html-italic">t</span>-test and one-way ANOVA were employed to compare multiple groups.</p>
Full article ">Figure 4
<p>Oxymatrine inhibited the expression of YY1 and the secretion of inflammatory factors. (<b>A</b>) Western blot analysis of YY1 levels following the administration of lipopolysaccharide (LPS) and oxymatrine in HK-2 cells. The signal densities of YY1 were normalized to that of β-actin. (<b>B</b>) Western blot analysis of YY1 levels after the administration of LPS and oxymatrine in HMC cells. The signal densities of YY1 were normalized to that of β-actin. (<b>C</b>) The levels of IL-6, IL-1β, and TNF-α in the culture medium of THP-1 cells. (<b>D</b>) The levels of IL-6, IL-1β, and TNF-α in the culture medium of HK-2 and HMC cells. (<b>E</b>) The levels of IL-6, IL-1β, and TNF-α in the culture medium of HK-2, HMC, and THP-1 cells. Each bar represents the mean ± SEM. * <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; one-way ANOVA was used to compare multiple groups.</p>
Full article ">Figure 5
<p>YY1 promotes inflammatory factor secretion in LN. (<b>A</b>) Western blot analysis of YY1 levels following the administration of LPS in HK-2 cells. The signal densities of YY1 were normalized to that of β-actin. (<b>B</b>) Western blot analysis of YY1 levels following the administration of LPS in HMC cells. The signal densities of YY1 were normalized to that of β-actin. (<b>C</b>) The relative activity of IL-6, IL-1β, and TNF-α in HK-2 cells was measured by luciferase assay. (<b>D</b>) The relative activity of IL-6, IL-1β, and TNF-α in HMC cells was measured by luciferase assay. (<b>E</b>) The levels of IL-6, IL-1β, and TNF-α in the culture medium of HK-2 cells after YY1 overexpression. (<b>F</b>) The levels of IL-6, IL-1β, and TNF-α in the culture medium of HMC cells after YY1 overexpression. (<b>G</b>) Western blot analysis of YY1 levels following YY1 knockdown in HK-2 cells. The signal densities of YY1 were normalized to that of β-actin. (<b>H</b>) Western blot analysis of YY1 levels following YY1 knockdown in HMC cells. The signal densities of YY1 were normalized to that of β-actin. (<b>I</b>) Cell colony formation assays of HK-2 and HMC cells after YY1 knockdown. (<b>J</b>) The levels of IL-6, IL-1β, and TNF-α in the culture medium of HK-2 cells after YY1 knockdown. (<b>K</b>) The levels of IL-6, IL-1β, and TNF-α in the culture medium of HMC cells after YY1 knockdown. Each bar represents the mean ± SEM. ns, no significance, * <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; <span class="html-italic">t</span>-test and one-way ANOVA were used to compare multiple groups.</p>
Full article ">Figure 6
<p>The effect of oxymatrine on LN is attributed to the inhibition of YY1. (<b>A</b>) Chemical structure of oxymatrine. (<b>B</b>) Homology modeling of YY1. (<b>C</b>) Ramachandran plots were utilized to evaluate the YY1 model. (<b>D</b>) Representative images illustrating the binding mode of oxymatrine and YY1. (<b>E</b>) Sensorgrams of oxymatrine in the SPR assay at various concentrations. (<b>F</b>) Levels of IL-6, IL-1β, and TNF-α in the culture medium of HK-2 cells following YY1 knockdown. (<b>G</b>) Levels of IL-6, IL-1β, and TNF-α in the culture medium of HMC cells following YY1 knockdown. (<b>H</b>) Levels of IL-6, IL-1β, and TNF-α in the culture medium of HK-2 cells after YY1 overexpression. (<b>I</b>) Levels of IL-6, IL-1β, and TNF-α in the culture medium of HMC cells after YY1 overexpression. Each bar represents the mean ± SEM. ns, no significance, ** <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; one-way ANOVA was used to compare multiple groups.</p>
Full article ">
9 pages, 592 KiB  
Article
Percutaneous Ultrasound-Guided Renal Biopsy in Greek Children: 15 Years of Experience at a Single Center
by John Dotis, Antonia Kondou, Vasiliki Karava, Pavlos Siolos, Vivian Georgopoulou, George Liapis, Maria Stamou, Fotios Papachristou and Nikoleta Printza
Pediatr. Rep. 2024, 16(4), 974-982; https://doi.org/10.3390/pediatric16040083 - 8 Nov 2024
Viewed by 647
Abstract
Background: Percutaneous ultrasound-guided renal biopsy (PRB) is a key element for diagnosis and management of several renal pathologies. We aimed to lay out the experience of our pediatric nephrology unit performing PRBs. The rationale and findings of these biopsies, safety issues and considerations [...] Read more.
Background: Percutaneous ultrasound-guided renal biopsy (PRB) is a key element for diagnosis and management of several renal pathologies. We aimed to lay out the experience of our pediatric nephrology unit performing PRBs. The rationale and findings of these biopsies, safety issues and considerations of the extracted data are going to be analyzed. Methods: A retrospective study was conducted from 2008 to 2023 based on the review of the medical records of pediatric patients who underwent PRBs. In total, 216 kidney biopsies in 206 patients were performed: 115 (53.2%) during the 2008–2015 period and 101 (46.8%) during the 2016–2023 period. Results: The most frequent clinical indication for PRBs was nephritic syndrome followed by nephrotic syndrome, observed in 84 (40.8%) and 72 (34.9%) patients, respectively. The predominant diagnosis was minimal change disease (MCD) (23.3%), followed by focal segmental glomerulosclerosis (FSGS) (15%) equal to lupus nephritis (LN) (15%), and immunoglobulin A nepropathy (10.2%). Minor complications, such as subcapsular hematomas were observed in approximately 15% of patients while no therapeutic intervention was needed. Conclusions: This report is the first review of pathohistological data covering a pediatric population over a 15-year period in Greece and one of the largest in southeastern Europe, especially in the Balkan region. The main indication for a PRB was nephritic syndrome; however, MCD was the main histological diagnosis. This study emphasis the fact that PRBs constitute a safe and reliable method of diagnostic approach to kidney diseases in childhood and offers important information on therapeutic approaches as well as the prognosis of these patients. Full article
Show Figures

Figure 1

Figure 1
<p>Differences of renal biopsy diagnosis based on age group. MCD minimal change disease, FSGS focal segmental glomerulosclerosis, IgAN immunoglobulin A nephropathy, LN lupus nephritis.</p>
Full article ">Figure 2
<p>Distribution of renal biopsy diagnoses between the two study periods in patients presented with isolated hematuria. IgAN immunoglobulin A nephropathy.</p>
Full article ">
13 pages, 3170 KiB  
Article
Neuropilin-1 as a Key Molecule for Renal Recovery in Lupus Nephritis: Insights from an NZB/W F1 Mouse Model
by Sebastian Sandoval, Cristina Solé, Blanca Joseph-Mullol, Maria Royo, Teresa Moliné, Alejandra Gabaldón and Josefina Cortés-Hernández
Int. J. Mol. Sci. 2024, 25(21), 11364; https://doi.org/10.3390/ijms252111364 - 22 Oct 2024
Viewed by 732
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease affecting multiple organs, with lupus nephritis (LN) occurring in 40–50% of SLE patients. Despite advances in diagnosis and treatment, LN remains a major cause of morbidity and mortality, with 10–20% of patients progressing to end-stage [...] Read more.
Systemic lupus erythematosus (SLE) is an autoimmune disease affecting multiple organs, with lupus nephritis (LN) occurring in 40–50% of SLE patients. Despite advances in diagnosis and treatment, LN remains a major cause of morbidity and mortality, with 10–20% of patients progressing to end-stage renal disease (ESRD). While knowledge of LN’s pathogenesis has improved, mechanisms of renal recovery are still largely unexplored. Neuropilin-1 (NRP-1), a transmembrane receptor expressed in renal tissue, has emerged as a promising biomarker for assessing renal recovery in LN. This study evaluates and correlates longitudinal levels of NRP-1 with kidney histology using an NZB/W F1 mouse model of LN. A total of 30 mice were used, with 15 receiving intravenous cyclophosphamide (CYC) and 15 being untreated. NRP-1 levels were measured in urine and serum, and kidney samples were taken from a subgroup of mice for histological evaluation. The results demonstrated a progressive increase in renal and urinary NRP-1 expression, particularly notable at weeks 26 and 32. Urinary NRP-1 levels above 34.40 ng/mL were strong predictors of favorable renal response, showing 100% sensitivity and 88% specificity. These findings indicate a robust correlation between urinary NRP-1 levels and renal histological recovery, underscoring the potential of NRP-1 as a valuable biomarker for assessing renal response in LN. This study demonstrates that renal production of NRP-1 is linked to histological recovery and establishes a foundation for future research into the role of NRP-1 in lupus kidney recovery. Full article
Show Figures

Figure 1

Figure 1
<p>The ameliorative effect of CYC on disease progression in NZB/W F1 mice. (<b>A</b>) Kaplan–Meier survival curve for NZB/W F1 mice treated with cyclophosphamide or saline (CYC-treated and non-treated groups, respectively). The log-rank test was used for survival analysis, with <span class="html-italic">p</span>-values ≤ 0.05 considered statistically significant. (<b>B</b>) Serum and urine biomarkers were measured to assess the progression of LN in both treated and non-treated mouse groups. Serum autoantibodies to double-stranded DNA (anti-dsDNA) and immunoglobulin (IgG) titers, as well as the urine protein/creatinine ratio (µg/mg), were determined using enzyme-linked immunosorbent assay (ELISA). One-way ANOVA followed by Bonferroni’s test and Student’s <span class="html-italic">t</span>-test were used to compare biomarker concentrations between the non-treated and CYC-treated groups. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Analysis of kidney tissue from untreated and CYC-treated groups. (<b>A</b>) Renal tissue was subjected to hematoxylin and eosin staining as well as immunofluorescence analysis for IgG and complement component C3. Cell nuclei were stained with DAPI (blue), while IgG and C3 proteins were labeled with FITC (green). Scale bar = 50 µm. Average scores for evaluation were obtained from the Vall d’Hebron pathology group to assess the percentage of glomeruli (<b>B</b>) and the staining intensity for IgG (<b>C</b>) and C3 (<b>D</b>) in renal tissue. One-way ANOVA followed by Bonferroni’s test and Student’s <span class="html-italic">t</span>-test were used to compare biomarker concentrations between groups. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Longitudinal analysis of NRP-1 protein levels during nephritis progression in NZB/W F1 mice. (<b>A</b>) Immunofluorescence staining for NRP-1 was performed on kidney tissue samples from both non-treated and CYC-treated groups at weeks 20, 26, 32, and at the study endpoint. Cell nuclei were stained with DAPI (blue), and NRP-1 was labeled with FITC (green). Scale bar = 50 µm. The intensity of NRP-1 immunofluorescence was quantified using the ImageJ Fiji software (version 1.45). Statistical comparisons between groups were performed using Student’s <span class="html-italic">t</span>-test. *** <span class="html-italic">p</span> &lt; 0.0001, **** <span class="html-italic">p</span> &lt; 0.00001. (<b>B</b>) Serum and urine levels of NRP-1 were measured at weeks 20, 26, 32, and at the study endpoint in both CYC-treated and non-treated NZB/W F1 mice groups. One-way ANOVA followed by Bonferroni’s post hoc test and Student’s <span class="html-italic">t</span>-test were used to compare NRP-1 levels between urine and serum at each time point. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Correlation between urinary NRP-1 levels (uNRP-1) and anti-dsDNA and IgG levels, protein/creatinine ratio, and mesangial proliferation. Spearman’s rank-order correlation was utilized to assess the relationships between these parameters, with significance levels and correlation coefficients indicated in each graph.</p>
Full article ">Figure 5
<p>Urinary NRP-1 levels as a potential biomarker for monitoring renal recovery. (<b>A</b>) Receiver operating characteristic (ROC) curve analysis of uNRP-1 was conducted using an optimal binary logistic regression model, combining data from both cohorts, to distinguish between renal recovery and non-recovery in NZB/W F1 mice (<span class="html-italic">n</span> = 11 for renal recovery, <span class="html-italic">n</span> = 19 for non-recovery). The area under the ROC curve (AUC) is provided. (<b>B</b>) Longitudinal analysis of uNRP-1 levels during the progression of lupus nephritis in NZB/W F1 mice was performed, categorizing mice based on whether they achieved renal recovery. Renal recovery was determined through histological analysis of kidney tissue, with mice exhibiting more than 40% mesangial proliferation being classified into the non-recovery group. Throughout the study period, mice in the renal recovery group consistently maintained uNRP-1 levels above 34.40 ng/mL.</p>
Full article ">
24 pages, 2418 KiB  
Review
Epitope Spreading in Immune-Mediated Glomerulonephritis: The Expanding Target
by Camillo Tancredi Strizzi, Martina Ambrogio, Francesca Zanoni, Bibiana Bonerba, Maria Elena Bracaccia, Giuseppe Grandaliano and Francesco Pesce
Int. J. Mol. Sci. 2024, 25(20), 11096; https://doi.org/10.3390/ijms252011096 - 16 Oct 2024
Viewed by 1372
Abstract
Epitope spreading is a critical mechanism driving the progression of autoimmune glomerulonephritis. This phenomenon, where immune responses broaden from a single epitope to encompass additional targets, contributes to the complexity and severity of diseases such as membranous nephropathy (MN), lupus nephritis (LN), and [...] Read more.
Epitope spreading is a critical mechanism driving the progression of autoimmune glomerulonephritis. This phenomenon, where immune responses broaden from a single epitope to encompass additional targets, contributes to the complexity and severity of diseases such as membranous nephropathy (MN), lupus nephritis (LN), and ANCA-associated vasculitis (AAV). In MN, intramolecular spreading within the phospholipase A2 receptor correlates with a worse prognosis, while LN exemplifies both intra- and intermolecular spreading, exacerbating renal involvement. Similarly, ANCA reactivity in AAV highlights the destructive potential of epitope diversification. Understanding these immunological cascades reveals therapeutic opportunities—targeting early epitope spreading could curb disease progression. Despite promising insights, the clinical utility of epitope spreading as a prognostic tool remains debated. This review provides a complete overview of the current evidence, exploring the dual-edged nature of epitope spreading, the intricate immune mechanisms behind it, and its therapeutic implications. By elucidating these dynamics, we aim to pave the way for more precise, targeted interventions in autoimmune glomerular diseases. Full article
(This article belongs to the Special Issue Molecular Advances in Glomerular Diseases)
Show Figures

Figure 1

Figure 1
<p><b>Intramolecular epitope spreading in PLA2R-associated membranous nephropathy.</b> The progression of intramolecular epitope spreading in PLA2R-associated membranous nephropathy (MN). Initially, autoantibodies target the CysR domain of the PLA2R receptor on podocytes. As the disease advances, antibody recognition extends to additional domains, including CTLD1, CTLD7, and CTLD8, correlating with an increase in antibody titers and disease severity. This spreading drives podocyte injury, foot process effacement, and disruption of the glomerular filtration barrier, resulting in proteinuria. External immune triggers may accelerate this process, worsening clinical outcomes and reducing the likelihood of remission. (Created in <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 2
<p><b>Epitope spreading in lupus nephritis.</b> This diagram illustrates the process of epitope spreading in lupus nephritis. Early in the disease, nucleosome–autoantibody complexes form in the mesangium, causing mild kidney damage. As the disease progresses, epitope spreading occurs, leading to the production of additional autoantibodies targeting histone proteins and other glomerular autoantigens, such as snRNP and C1q. This results in immune complex deposition in the subendothelial and subepithelial spaces, contributing to more severe forms of lupus nephritis (Class III/IV). This process not only exacerbates autoimmunity but also contributes to the progression and severity of the diseases, significantly contributing to the heterogeneity and severity of renal involvement in lupus patients. Furthermore, kidney tertiary lymphoid structures (TLSs) can promote a localized immune response against specific autoantigens overexpressed in the inflamed tissue, in association with epitope spreading, higher disease activity, and poor treatment response. (Created in <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 3
<p><b>Epitope spreading in ANCA-associated vasculitis.</b> The image illustrates the process of epitope spreading in ANCA-associated vasculitis (AAV). Initially, PR3–ANCA and MPO–ANCA antibodies target specific epitopes on PR3 and MPO, respectively. As the disease progresses, intramolecular epitope spreading occurs, leading to the recognition of new epitopes. This spreading contributes to increased vascular injury and inflammation, playing a crucial role in disease progression and relapse. (Created in <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">
10 pages, 1348 KiB  
Brief Report
Urine Extracellular Vesicles Size Subsets as Lupus Nephritis Biomarkers
by Itze C. Navarro-Hernandez, Raúl F. Reyes-Huerta, Mariana Cañez-Hernández, Jiram Torres-Ruiz, Daniel A. Carrillo-Vázquez, Laura P. Whittall-García, David E. Meza-Sánchez, Guillermo Juárez-Vega, Diana Gómez-Martin, José M. Hernández-Hernández and José L. Maravillas-Montero
Diagnostics 2024, 14(20), 2271; https://doi.org/10.3390/diagnostics14202271 - 12 Oct 2024
Viewed by 793
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder that often leads to kidney injury, known as lupus nephritis (LN). Although renal biopsy is the primary way to diagnose LN, it is invasive and not practical for regular monitoring. As an alternative, several groups [...] Read more.
Systemic lupus erythematosus (SLE) is an autoimmune disorder that often leads to kidney injury, known as lupus nephritis (LN). Although renal biopsy is the primary way to diagnose LN, it is invasive and not practical for regular monitoring. As an alternative, several groups have proposed urinary extracellular vesicles (uEVs) as potential biomarkers for LN, as recent studies have shown their significance in reflecting kidney-related diseases. As a result, we developed a flow cytometry approach that allowed us to determine that LN patients exhibited a significantly higher total uEV concentration compared to SLE patients without kidney involvement. Additionally, an analysis of different-sized uEV subsets revealed that microvesicles ranging from 0.3 to 0.5 μm showed the most promise for distinguishing LN. These findings indicate that evaluating uEV concentration and size distribution could be a valuable diagnostic and monitoring tool for LN, pending further validation in more comprehensive studies. Full article
(This article belongs to the Special Issue Updates in the Diagnosis and Management of Autoimmune Diseases)
Show Figures

Figure 1

Figure 1
<p>uEVs display no differences in size distribution but exhibit increased concentration in LN. (<b>A</b>) Gates for each bead size (upper panel) employed as a reference to define the relative size of purified uEVs in patient samples (lower panel, showing a representative contour plot). FSC—forward scatter (size); SSC—side scatter (granularity). (<b>B</b>) Size distribution plot displaying normalized mean frequencies of each uEV size subset in SLE vs. LN patients. (<b>C</b>) Total uEV concentration in SLE vs. LN patients. The data were analyzed using the Mann–Whitney U test. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>uEV levels correlate with different clinical features in SLE/LN patients. Correlation matrix of the calculated Spearman coefficient between uEV concentrations of different sizes (left side) and clinical characteristics of SLE/LN patients (upper side). The sidebar color scale (right side) indicates the magnitude of a positive correlation (red) or a negative correlation (blue). The size of each circle represents the correlation coefficient enclosed. All the displayed correlations in the plot are significant (<span class="html-italic">p</span> &lt; 0.05), besides the non-significant correlations that are represented with empty white squares.</p>
Full article ">Figure 3
<p>ROC curves of uEV concentrations for discrimination of LN patients and non-kidney affected SLE patients. The values of each AUC and <span class="html-italic">p</span>-values are depicted. <span class="html-italic">p</span>-values &lt; 0.05 were considered significant. SLE patients (<span class="html-italic">n</span> = 11) and LN (<span class="html-italic">n</span> = 15).</p>
Full article ">
20 pages, 2383 KiB  
Review
The Role of Antioxidant Transcription Factor Nrf2 and Its Activating Compounds in Systemic Lupus Erythematosus
by Lu Liu, Karina de Leeuw, Harry van Goor and Johanna Westra
Antioxidants 2024, 13(10), 1224; https://doi.org/10.3390/antiox13101224 - 11 Oct 2024
Viewed by 1228
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease in which kidney involvement, so-called lupus nephritis (LN), is common and one of the most severe manifestations. Oxidative stress (OS) may play a role in the pathogenesis of LN through the exacerbation of inflammation [...] Read more.
Systemic lupus erythematosus (SLE) is a complex autoimmune disease in which kidney involvement, so-called lupus nephritis (LN), is common and one of the most severe manifestations. Oxidative stress (OS) may play a role in the pathogenesis of LN through the exacerbation of inflammation and immune cell dysfunction/dysregulation. Nuclear factor erythroid 2-related factor 2 (Nrf2), also known as nuclear factor erythroid-derived 2-like 2, is a transcription factor that in humans is encoded by the NFE2L2 gene and is regarded as a central regulator of the antioxidative response. Nrf2-activating compounds have been shown to alleviate oxidative stress in cells and tissues of lupus-prone mice. Although the precise mechanisms of Nrf2 activation on the immune system in SLE remain to be elucidated, Nrf2-activating compounds are considered novel therapeutical options to suppress OS and thereby might alleviate disease activity in SLE, especially in LN. This review therefore summarizes the role of the Nrf2 signaling pathway in the pathogenesis of SLE with LN and describes compounds modulating this pathway as potential additional clinical interventions. Full article
(This article belongs to the Special Issue Oxidative Stress in Renal Health)
Show Figures

Figure 1

Figure 1
<p>The biological functions of the Nrf2 pathway and interaction with oxidative stress.</p>
Full article ">Figure 2
<p>The depiction of the crosstalk of the Nrf2 and NF-κΒ in LN. ↑, upregulation; ↓, downregulation.</p>
Full article ">Figure 3
<p>Current Nrf2 activators described in SLE.</p>
Full article ">
Back to TopTop