[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (5,975)

Search Parameters:
Keywords = immunohistochemistry

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
11 pages, 732 KiB  
Article
Assessing Influence of Mismatch Repair Mutations on Survival in Patients After Resection of Pancreatic Ductal and Periampullary Adenocarcinoma
by Elizabeth Prezioso, Eleanor Mancheski, Kylee Shivok, Zachary Kaplan, Wilbur Bowne, Aditi Jain, Harish Lavu, Charles J. Yeo and Avinoam Nevler
J. Clin. Med. 2024, 13(20), 6185; https://doi.org/10.3390/jcm13206185 (registering DOI) - 17 Oct 2024
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related deaths in the United States. Previous studies have indicated that microsatellite instability and deficient mismatch repair (MMR) may be associated with improved survival in patients with pancreatic cancer. Here, we [...] Read more.
Background: Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related deaths in the United States. Previous studies have indicated that microsatellite instability and deficient mismatch repair (MMR) may be associated with improved survival in patients with pancreatic cancer. Here, we aim to investigate the impact of deficient MMR (dMMR) status on oncologic outcomes in patients after resection of PDAC and periampullary adenocarcinoma. Methods: This is a single-institution, retrospective study based on a prospectively maintained database. Pancreatic ductal adenocarcinoma (N = 342) and periampullary adenocarcinoma patients (N = 76) who underwent pancreatic resection surgery between 2016 and 2021 were included. Immunohistochemistry staining results of MMR proteins and next-generation sequencing data were recorded. Cancer-type dependent Cox regression analyses were performed to assess overall and disease-free survival, which was complemented with a 1:2 propensity-score matching for each of the cancer types in order to compare oncologic outcomes. Results: A total of 418 pancreatic cancer patients were included in the analysis. Fifteen patients (3.5%) were diagnosed as dMMR (PDAC N = 7 and periampullary adenocarcinoma N = 8). Cox regression modeling of dMMR status interaction with TNM staging and cancer type revealed that dMMR status strongly improves overall survival (p < 0.05). After propensity-score matching, Cox regression identified dMMR status as a significant marker of improved overall survival (HR = 0.27, 95%CI 0.09–0.88, p = 0.029). Conclusions: Overall, our findings suggest that dMMR status is associated with markedly improved survival outcomes in patients after resection of pancreatic and periampullary cancer. Future large-scale studies are needed to further validate this finding. Full article
(This article belongs to the Special Issue Targeted Treatment of Pancreatic Cancer)
Show Figures

Figure 1

Figure 1
<p>Cox regression models for (<b>A</b>) overall survival and (<b>B</b>) disease-free survival (N = 418). dMMR—deficient MMR; pMMR—proficient MMR.</p>
Full article ">Figure 2
<p>Propensity-score-matched (1:2) and weighted Cox regression model for overall survival analysis in pancreatic ductal adenocarcinoma (<b>A</b>) and periampullary cancer patients (<b>B</b>), assessing the impact of deficient MMR on the two cancer types (dMMR = 15/pMMR = 30).</p>
Full article ">
20 pages, 4472 KiB  
Article
Loureirin B Reduces Insulin Resistance and Chronic Inflammation in a Rat Model of Polycystic Ovary Syndrome by Upregulating GPR120 and Activating the LKB1/AMPK Signaling Pathway
by Jing Wang, Zheng Huang, Zhiyong Cao, Yehao Luo, Yueting Liu, Huilu Cao, Xiusong Tang and Gang Fang
Int. J. Mol. Sci. 2024, 25(20), 11146; https://doi.org/10.3390/ijms252011146 - 17 Oct 2024
Abstract
Polycystic ovary yndrome (PCOS) is a common metabolic disorder in women, which is usually associated with insulin resistance (IR) and chronic inflammation. Loureirin B (LrB) can effectively improve insulin resistance and alleviate chronic inflammation, and in order to investigate the therapeutic effect of [...] Read more.
Polycystic ovary yndrome (PCOS) is a common metabolic disorder in women, which is usually associated with insulin resistance (IR) and chronic inflammation. Loureirin B (LrB) can effectively improve insulin resistance and alleviate chronic inflammation, and in order to investigate the therapeutic effect of LrB on polycystic ovary syndrome with insulin resistance (PCOS-IR), we conducted animal experiments. A PCOS-IR rat model was established by feeding a high-fat diet combined with letrozole (1 mg/kg·d for 21 days). The rats were treated with the GPR120 agonists TUG-891 and LrB for 4 weeks. Biochemical parameters (fasting blood glucose, total cholesterol, triglycerides, high- and low-density lipoprotein), hormone levels (serum insulin, E2, T, LH, and FSH), and inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-18) were analyzed. Histopathological analyses of ovaries were performed using hematoxylin/eosin (H&E) staining. Real-time PCR and western blotting were used to assess GPR120, NLRP3, and caspase-1 expression in ovaries, and immunohistochemistry was used to evaluate LKB1 and AMPK protein expression. LrB reduced body weight, Lee’s index, ovarian index, ovarian area, and volume in PCOS-IR rats. It lowered fasting blood glucose, serum insulin, and HOMA-IR. LrB decreased total serum cholesterol, triglyceride, and LDL levels and increased HDL levels. It reduced serum T, LH, and LH/FSH and raised serum E2 and FSH levels. LrB downregulated the mRNA and protein expression levels of NLRP3 and Caspase-1, increased the protein and mRNA expression levels of GPR120 in rat ovaries, and increased LKB1 and AMPK protein expression in ovaries, ameliorating ovarian histopathological changes in PCOS-IR rats. Taken together, LrB upregulated GPR120, LKB1, and AMPK protein expression, downregulated NLRP3 and Caspase-1 protein expression, reduced insulin resistance and chronic inflammation, and ameliorated histopathological changes in ovarian tissues in PCOS rats, suggesting its potential as a treatment for PCOS. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Therapeutic effect of LrB on PCOS-IR rats.</p>
Full article ">Figure 2
<p>Effect of LrB on body weight and Lee’s index in PCOS-IR rats. The data are mean ± SDs, n = 10. Body weights of rats at (<b>A</b>) week 0 of treatment, (<b>B</b>) week 1, (<b>C</b>) week 2, (<b>D</b>) week 3, (<b>E</b>) week 4. (<b>F</b>) Lee’s indices of the rats in each group after four weeks of treatment. In comparison with the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; in comparison with the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05; ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 3
<p>Effect of LrB on the ovarian area, ovarian volume, and ovarian index in PCOS-IR rats. The values are means ± SDs; n = 10. (<b>A</b>) Ovarian area at four weeks of treatment. (<b>B</b>) Ovarian volume at four weeks. (<b>C</b>) Ovarian indices at four weeks. In comparison with the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; in comparison with the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05; ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 4
<p>Vaginal cytology images (magnification 10×, 40×). LrB improved the estrous cycle in PCOS-IR rats.</p>
Full article ">Figure 5
<p>Effect of LrB on lipid metabolic activity in PCOS-IR rats. The values are means ± SDs; n = 10. (<b>A</b>) TC levels at four weeks of treatment. (<b>B</b>) TG levels at four weeks. (<b>C</b>) LDL levels at four weeks. (<b>D</b>) HDL levels at four weeks. Relative to the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; relative to the PCOS-IR group, ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 6
<p>Effect of LrB on fasting blood glucose and serum insulin in PCOS-IR rats; values are presented as the means ± SDs, n = 10. (<b>A</b>) INS levels at four weeks of treatment. (<b>B</b>) FBG levels at four weeks. (<b>C</b>) HOMA-IR at four weeks. Relative to the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; relative to the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05; ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 7
<p>Effects of LrB on hormone levels in PCOS-IR rats; values are means ± SDs; n = 10. (<b>A</b>) LH levels at four weeks of treatment. (<b>B</b>) T levels at four weeks. (<b>C</b>) LH/FSH ratio at four weeks. (<b>D</b>) FSH levels at four weeks. (<b>E</b>) E2 levels at four weeks. Relative to the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; relative to the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05; ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 8
<p>LrB alleviates the serum inflammatory response in PCOS-IR rats. Values are means ± SDs; n = 10. (<b>A</b>) Serum TNF-α content at four weeks of treatment. (<b>B</b>) Serum IL-1β at four weeks. (<b>C</b>) Serum IL-6 at four weeks. (<b>D</b>) Serum IL-18 at four weeks. Relative to the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; relative to the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05, ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 9
<p>Effects of LrB treatment on morphological changes in ovarian tissue (magnification 4×, 10×). Representative H&amp;E-stained ovarian tissue sections after four weeks of treatment. C, cystic follicles; CL, corpus luteum; ANF, antral follicles; ATF, atretic follicles.</p>
Full article ">Figure 10
<p>Effects of LrB on GPR120 expression in PCOS-IR rat ovaries. Data are means ± SDs; n = 3. (<b>A</b>) Western blot analysis of GPR120 and GAPDH in ovarian tissues at four weeks of treatment. (<b>B</b>) Quantitative analysis of GPR120 levels in ovarian tissues at four weeks of treatment. (<b>C</b>) Expression levels of GPR120 mRNA in ovarian tissues at four weeks of treatment. Relative to the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; relative to the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05; ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 11
<p>Effects of LrB on NLRP3 and Caspase-1 levels in the ovaries of PCOS-IR rats. The values are means ± SDs; n = 3. (<b>A</b>) Western blot analysis of NLRP3 and GAPDH in ovarian tissues at four weeks of treatment. (<b>B</b>) Quantitative analysis of NLRP3 levels in ovarian tissues at four weeks of treatment. (<b>C</b>) Expression levels of NLRP3 mRNA in ovarian tissues at four weeks of treatment. (<b>D</b>) Western blot analysis of Caspase-1 and GAPDH in ovarian tissues at four weeks of treatment. (<b>E</b>) Quantitative analysis of Caspase-1 levels in ovarian tissues at four weeks of treatment. (<b>F</b>) Expression levels of Caspase-1 mRNA in ovarian tissues at four weeks of treatment. Relative to the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; relative to the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05; ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 12
<p>The influence of LrB on expression of LKB1 and AMPK in the ovaries of PCOS-IR rats. The values are means ± SDs; n = 5. (<b>A</b>) Expression of AMPK in the ovarian tissues of each group at four weeks of treatment (magnification 10×, 40×). (<b>B</b>) Expression of LKB1 in the ovarian tissues of each group at four weeks of treatment (magnification 10×, 40×). (<b>C</b>) Positive expression of AMPK was analyzed. (<b>D</b>) Positive expression of LKB1 was analyzed. Relative to the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; relative to the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05; ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 12 Cont.
<p>The influence of LrB on expression of LKB1 and AMPK in the ovaries of PCOS-IR rats. The values are means ± SDs; n = 5. (<b>A</b>) Expression of AMPK in the ovarian tissues of each group at four weeks of treatment (magnification 10×, 40×). (<b>B</b>) Expression of LKB1 in the ovarian tissues of each group at four weeks of treatment (magnification 10×, 40×). (<b>C</b>) Positive expression of AMPK was analyzed. (<b>D</b>) Positive expression of LKB1 was analyzed. Relative to the normal group, <sup>##</sup> <span class="html-italic">p &lt;</span> 0.01; relative to the PCOS-IR group, * <span class="html-italic">p &lt;</span> 0.05; ** <span class="html-italic">p &lt;</span> 0.01.</p>
Full article ">Figure 13
<p>Animal grouping and handling procedures.</p>
Full article ">
18 pages, 901 KiB  
Systematic Review
Characterization of the Joint Microenvironment in Osteoarthritic Joints for In Vitro Strategies for MSC-Based Therapies: A Systematic Review
by Aline Silvestrini da Silva, Fernanda Campos Hertel, Fabrício Luciani Valente, Fabiana Azevedo Voorwald, Andrea Pacheco Batista Borges, Adriano de Paula Sabino, Rodrigo Viana Sepulveda and Emily Correna Carlo Reis
Appl. Biosci. 2024, 3(4), 450-467; https://doi.org/10.3390/applbiosci3040029 (registering DOI) - 17 Oct 2024
Abstract
Osteoarthritis is a joint disease that causes pain, stiffness, and reduced joint function because the protective cushioning inside the joints, called cartilage, gradually wears away. This condition is caused by various factors and complex processes in the joint’s environment, involving different types of [...] Read more.
Osteoarthritis is a joint disease that causes pain, stiffness, and reduced joint function because the protective cushioning inside the joints, called cartilage, gradually wears away. This condition is caused by various factors and complex processes in the joint’s environment, involving different types of cells producing factors that can either maintain the joint health or contribute to osteoarthritis. This study aimed to understand the factors influencing both healthy and diseased joints in DDD strategies for the in vitro preconditioning of MSCs. An electronic search in the PubMed, Scopus, and Web of Science databases was carried out using the terms (cartilage OR chondr*) AND (repair OR regeneration OR healing) AND (niche OR microenvironment)) AND (“growth factor” OR GF OR cytokine). Researchers used various methods, including macroscopic examinations, histology, immunohistochemistry, and microCT. Molecules associated with joint inflammation were identified, like macrophage markers, MMP-13, TNF, apoptotic markers, and interleukins. Chondrogenesis-related factors such as aggrecan GAG, collagen type II, and TGF beta family were also identified. This study suggests that balancing certain molecules and ensuring the survival of joint chondrocytes could be crucial in improving the condition of osteoarthritic joints, emphasizing the importance of chondrocyte survival and activity. Future preconditioning methods for MSC- and EV-based therapies can find suitable strategies in the described microenvironments to explore co-culture systems and soluble or extracellular matrix factors. Full article
(This article belongs to the Special Issue Anatomy and Regenerative Medicine: From Methods to Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Flowchart of the systematic review mechanism.</p>
Full article ">Figure 2
<p>Risk of bias in assessing the methodological quality of the 20 articles included in this systematic review.</p>
Full article ">
14 pages, 8177 KiB  
Article
The Protective Role of Intermedin in Contrast-Induced Acute Kidney Injury: Enhancing Peritubular Capillary Endothelial Cell Adhesion and Integrity Through the cAMP/Rac1 Pathway
by Tingting Gao, Ruiyuan Gu, Heng Wang, Lizheng Li, Bojin Zhang, Jie Hu, Qinqin Tian, Runze Chang, Ruijing Zhang, Guoping Zheng and Honglin Dong
Int. J. Mol. Sci. 2024, 25(20), 11110; https://doi.org/10.3390/ijms252011110 - 16 Oct 2024
Viewed by 239
Abstract
Contrast-induced acute kidney injury (CIAKI) is a common complication with limited treatments. Intermedin (IMD), a peptide belonging to the calcitonin gene-related peptide family, promotes vasodilation and endothelial stability, but its role in mitigating CIAKI remains unexplored. This study investigates the protective effects of [...] Read more.
Contrast-induced acute kidney injury (CIAKI) is a common complication with limited treatments. Intermedin (IMD), a peptide belonging to the calcitonin gene-related peptide family, promotes vasodilation and endothelial stability, but its role in mitigating CIAKI remains unexplored. This study investigates the protective effects of IMD in CIAKI, focusing on its mechanisms, particularly the cAMP/Rac1 signaling pathway. Human umbilical vein endothelial cells (HUVECs) were treated with iohexol to simulate kidney injury in vitro. The protective effects of IMD were assessed using CCK8 assay, flow cytometry, ELISA, and Western blotting. A CIAKI rat model was utilized to evaluate renal peritubular capillary endothelial cell injury and renal function through histopathology, immunohistochemistry, immunofluorescence, Western blotting, and transmission electron microscopy. In vitro, IMD significantly enhanced HUVEC viability and mitigated iohexol-induced toxicity by preserving intercellular adhesion junctions and activating the cAMP/Rac1 pathway, with Rac1 inhibition attenuating these protective effects. In vivo, CIAKI caused severe damage to peritubular capillary endothelial cell junctions, impairing renal function. IMD treatment markedly improved renal function, an effect negated by Rac1 inhibition. IMD protects against renal injury in CIAKI by activating the cAMP/Rac1 pathway, preserving peritubular capillary endothelial integrity and alleviating acute renal injury from contrast media. These findings suggest that IMD has therapeutic potential in CIAKI and highlight the cAMP/Rac1 pathway as a promising target for preventing contrast-induced acute kidney injury in at-risk patients, ultimately improving clinical outcomes. Full article
Show Figures

Figure 1

Figure 1
<p>IMD can antagonize damage to HUVEC viability and apoptosis induced by iohexol. (<b>A</b>) HUVECs were preincubated with IMD (0, 1, 10, 100 nmol/L) for 30 min and then treated with iohexol (10, 20, 40, 80 mgI/mL) for 12 h. A CCK-8 kit was used to test cell viability. (<b>B</b>) HUVECs were preincubated with IMD (10 nmol/L) for 30 min, then were treated with iohexol (40 mgI/mL) or iohexol (40 mgI/mL) +NSC23766(50 μM) for 12 h. Apoptosis was detected using flow cytometry. (<b>C</b>) The apoptosis rate in each group was quantified (<span class="html-italic">n</span> = 6). The results were analyzed using ANOVA, followed by Tukey’s multiple comparisons test for subgroup analysis. All data are expressed as mean ± SD, * <span class="html-italic">p</span> &lt; 0.05 compared to the control group, # <span class="html-italic">p</span> &lt; 0.05. For comparison between groups, ns: not significant.</p>
Full article ">Figure 2
<p>IMD can protect the adherens junction of HUVECs by activating the cAMP/Rac1 pathway. (<b>A</b>) Intermedin induces cAMP production in HUVECs. The concentration of cAMP was measured using ELISA, <span class="html-italic">n</span> = 4. (<b>B</b>,<b>C</b>) HUVECs were treated with 10 nmol of IMD and/or 40 mgI/mL iohexol or 50 μM NSC23766 for 12 h. Whole-cell lysates of HUVECs were collected for immunoblotting analysis of Rac1, VE-cadherin, and GAPDH. The results were analyzed using ANOVA, with Tukey’s multiple comparisons test applied for subgroup analysis. All data are expressed as mean ± SD, * <span class="html-italic">p</span> &lt; 0.05 compared to the control group, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05. For comparison between groups, ns: not significant.</p>
Full article ">Figure 3
<p>IMD attenuates renal injury in rat CIAKI models, and inhibition of Rac1 might abolish this protective effect. (<b>A</b>) Male SD rats were randomly divided into five groups (<span class="html-italic">n</span> = 6), and the rats in each group were treated as shown in the scheme. (<b>B</b>) Renal morphology (HE, magnification ×400) showed vacuolar degeneration of renal tubules (shown by red arrows) and dilatation of renal tubules (shown by green arrows). (<b>C</b>) Magnification of renal PAS staining (×400) showed the absence of brush edges of renal tubules, vacuolar degeneration, and dilatation of renal tubules. (<b>D</b>) Renal tubular injury score. (<b>E</b>) Serum creatinine levels for the indicated treatments. The results were analyzed using ANOVA, with Tukey’s multiple comparisons test applied for subgroup analysis. All data are expressed as mean ± SD, * <span class="html-italic">p</span> &lt; 0.05 compared to the control group, # <span class="html-italic">p</span> &lt; 0.05. For comparison between groups, ns: not significant.</p>
Full article ">Figure 4
<p>IMD activates the cAMP/Rac1 pathway and alleviates renal peritubular capillary injury in CIAKI rats. (<b>A</b>,<b>B</b>): The renal lysates of rats were collected and the expression of Rac1, VE-cadherin, VEGFR2, and GAPDH were detected using Western blotting. (<b>C</b>) Kidney sections were stained with CD34 immunohistochemical staining (magnification, ×400, scale 30 μm). (<b>D</b>) Kidney sections were stained with CD34 and ICAM1 immunofluorescence staining (magnification, ×200, scale 50 μm). (<b>E</b>) The average optical density of CD34 positive staining in each group was quantified (<span class="html-italic">n</span> = 6). (<b>E</b>) Quantification of the average optical density of ICAM1 immunofluorescence (<span class="html-italic">n</span> = 6). The data were analyzed using ANOVA, followed by Tukey’s multiple comparisons test for subgroup analysis. All data are expressed as mean ± SD, * <span class="html-italic">p</span> &lt; 0.05 compared to the control group, # <span class="html-italic">p</span> &lt; 0.05. For comparison between groups, ns: not significant.</p>
Full article ">Figure 5
<p>IMD protects the PTC endothelial barrier: a representative microphotograph of the ultrastructural changes of peritubular capillary endothelial cells, cell membrane discontinuity (shown by green arrows), and basement membrane fracture (shown by blue arrows) (original magnification, ×12,000; scale, 1 μm).</p>
Full article ">
14 pages, 2928 KiB  
Article
PEP-1–PIN1 Promotes Hippocampal Neuronal Cell Survival by Inhibiting Cellular ROS and MAPK Phosphorylation
by Jung Hwan Park, Min Jea Shin, Gi Soo Youn, Hyeon Ji Yeo, Eun Ji Yeo, Hyun Jung Kwon, Lee Re Lee, Na Yeon Kim, Su Yeon Kwon, Su Min Kim, Yong-Jun Cho, Sung Ho Lee, Hyo Young Jung, Dae Won Kim, Won Sik Eum and Soo Young Choi
Biomedicines 2024, 12(10), 2352; https://doi.org/10.3390/biomedicines12102352 (registering DOI) - 15 Oct 2024
Viewed by 435
Abstract
Background: The peptidyl-prolyl isomerase (PIN1) plays a vital role in cellular processes, including intracellular signaling and apoptosis. While oxidative stress is considered one of the primary mechanisms of pathogenesis in brain ischemic injury, the precise function of PIN1 in this disease remains [...] Read more.
Background: The peptidyl-prolyl isomerase (PIN1) plays a vital role in cellular processes, including intracellular signaling and apoptosis. While oxidative stress is considered one of the primary mechanisms of pathogenesis in brain ischemic injury, the precise function of PIN1 in this disease remains to be elucidated. Objective: We constructed a cell-permeable PEP-1–PIN1 fusion protein and investigated PIN1’s function in HT-22 hippocampal cells as well as in a brain ischemic injury gerbil model. Methods: Transduction of PEP-1–PIN1 into HT-22 cells and signaling pathways were determined by Western blot analysis. Intracellular reactive oxygen species (ROS) production and DNA damage was confirmed by DCF-DA and TUNEL staining. Cell viability was determined by MTT assay. Protective effects of PEP-1-PIN1 against ischemic injury were examined using immunohistochemistry. Results: PEP-1–PIN1, when transduced into HT-22 hippocampal cells, inhibited cell death in H2O2-treated cells and markedly reduced DNA fragmentation and ROS production. This fusion protein also reduced phosphorylation of mitogen-activated protein kinase (MAPK) and modulated expression levels of apoptosis-signaling proteins in HT-22 cells. Furthermore, PEP-1–PIN1 was distributed in gerbil hippocampus neuronal cells after passing through the blood–brain barrier (BBB) and significantly protected against neuronal cell death and also decreased activation of microglia and astrocytes in an ischemic injury gerbil model. Conclusions: These results indicate that PEP-1–PIN1 can inhibit ischemic brain injury by reducing cellular ROS levels and regulating MAPK and apoptosis-signaling pathways, suggesting that PIN1 plays a protective role in H2O2-treated HT-22 cells and ischemic injury gerbil model. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 1

Figure 1
<p>Construction of PEP-1–PIN1 and control PIN1 protein. Constructed map of PEP-1–PIN1 based on the pET-15b vector. PEP-1–PIN1 was designed to contain histidine, PEP-1-PTD and PIN1 (<b>A</b>). Purified PEP-1–PIN1 and control PIN1 were confirmed by Coomassie brilliant blue staining and Western blot analysis using anti-histidine antibody (<b>B</b>).</p>
Full article ">Figure 2
<p>Delivery of PEP-1–PIN1 into HT-22 cells. HT-22 cells were treated with PEP-1–PIN1 (0.5–5 μM) for 3 h (<b>A</b>) or PEP-1–PIN1 (5 μM) for different time periods (30–180 min) (<b>B</b>). The intracellular stability of delivered PEP-1–PIN1 into the cells. HT-22 cells were treated with PEP-1–PIN1 for 3 h and washed. The cells were then further incubated for 1 to 60 h (<b>C</b>) and delivered PEP-1–PIN1 was assessed by Western blotting. The intensity of the bands was measured by a densitometer. Data are represented as mean ± SEM (n = 3).</p>
Full article ">Figure 3
<p>Effects of delivered PEP-1–PIN1 against H<sub>2</sub>O<sub>2</sub>-induced cell death. HT-22 cells were treated with PEP-1–PIN1 (5 μM) for 3 h. The localization of delivered PEP-1–PIN1 was confirmed by fluorescence microscopy (<b>A</b>). Scale bar = 20 μm. Effect of delivered PEP-1–PIN1 against H<sub>2</sub>O<sub>2</sub>-induced cell viability. The cells were pretreated with PEP-1–PIN1 (0.5–5 μM) for 3 h and exposed to H<sub>2</sub>O<sub>2</sub> (1 mM) for 2 h. Cell viability was assessed by MTT assay (<b>B</b>). Data are represented as mean ± SEM (n = 3). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with H<sub>2</sub>O<sub>2</sub>-treated cells.</p>
Full article ">Figure 4
<p>Effects of delivered PEP-1–PIN1 against H<sub>2</sub>O<sub>2</sub>-induced ROS production and DNA damage. HT-22 cells were treated with PEP-1–PIN1 (5 μM) for 3 h before treatment with 1 mM H<sub>2</sub>O<sub>2</sub> for 1 h or 6 h. Intracellular ROS levels (<b>A</b>) and DNA damage (<b>B</b>) were determined by DCF-DA and TUNEL staining. Fluorescence intensity was quantified using an ELISA plate reader. Scale bar = 50 μm. Data are represented as mean ± SEM (n = 3). ** <span class="html-italic">p</span> &lt; 0.01 compared with H<sub>2</sub>O<sub>2</sub>-treated cells.</p>
Full article ">Figure 5
<p>Effects of delivered PEP-1–PIN1 against H<sub>2</sub>O<sub>2</sub>-induced MAPK and NF-κB expression in HT-22 cells. The cells were treated with PEP-1–PIN1 (5 μM) for 3 h before being exposed to H<sub>2</sub>O<sub>2</sub> (1 mM) for 60 min or 30 min, respectively. The expression levels of MAPKs (<b>A</b>) and NF-κB (<b>B</b>) were analyzed by Western blotting. The intensity of the bands was measured by a densitometer. Data are represented as mean ± SEM (n = 3). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with H<sub>2</sub>O<sub>2</sub> treated cells.</p>
Full article ">Figure 6
<p>Effects of delivered PEP-1–PIN1 against H<sub>2</sub>O<sub>2</sub>-induced Bax, Bcl-2, and p53 protein expression in HT-22 cells. Three-hour pretreatment of HT-22 cells with PEP-1–PIN1 (5 μM) was followed by treatments with H<sub>2</sub>O<sub>2</sub> (1 mM) for 120 min (Bcl-2), 240 min (Bax), and 360 min (p53). The expression levels of Bcl-2 and Bax (<b>A</b>) and p53 (<b>B</b>) were determined by Western blot analysis. The intensity of the bands was measured by a densitometer. Data are represented as mean ± SEM (n = 3). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with H<sub>2</sub>O<sub>2</sub> treated cells.</p>
Full article ">Figure 7
<p>The neuroprotective effects of delivered PEP-1–PIN1 against ischemic damage. Gerbils were treated with a single injection of PEP-1–PIN1 (2 mg/kg) and sacrificed after 7 days. Delivery of PEP-1–PIN1 into the CA1 region of the hippocampus was determined by anti-histidine immunohistochemistry. Scale bar = 400 μm. The hippocampus was stained with NeuN, CV, GFAP, Iba-1 and FJB in sham-, vehicle-, PEP-1–PIN1-, PIN1-, and PEP-1-treated gerbils after ischemic injury. The graphic shows the relative numerical analyses of CV, GFAP, Iba-1 and FJB positive neurons in the CA1 region. Scale bar = 400 and 50 μm. Each bar represents the mean ± SEM of ten mice. ** <span class="html-italic">p</span> &lt; 0.01, significant difference from the vehicle group.</p>
Full article ">
16 pages, 1471 KiB  
Article
Alteration in sB7-H4 Serum Levels and Placental Biomarker Expression after Therapeutic Plasma Exchange in Early-Onset Preeclampsia Patients
by Liyan Duan, Yuyang Ma, Beatrix Reisch, Elina Hadrovic, Pawel Mach, Rainer Kimmig, Michael Jahn, Angela Köninger, Antonella Iannaccone and Alexandra Gellhaus
Int. J. Mol. Sci. 2024, 25(20), 11082; https://doi.org/10.3390/ijms252011082 (registering DOI) - 15 Oct 2024
Viewed by 228
Abstract
Therapeutic plasma exchange (TPE) is a widely used treatment for numerous diseases including pregnancy-related conditions. Our prior study on 20 early-onset preeclampsia patients undergoing TPE revealed a significant extension in pregnancy duration and reduced serum levels of sFlt-1, sFlt-1/PlGF, and sEndoglin. Here, we [...] Read more.
Therapeutic plasma exchange (TPE) is a widely used treatment for numerous diseases including pregnancy-related conditions. Our prior study on 20 early-onset preeclampsia patients undergoing TPE revealed a significant extension in pregnancy duration and reduced serum levels of sFlt-1, sFlt-1/PlGF, and sEndoglin. Here, we investigated the impact of TPE on serum sB7-H4, an immunological checkpoint molecule, and placental proteins (Flt-1, Eng, B7-H4, iNOS, TNF-α) in TPE-treated early-onset preeclampsia patients (N = 12, 23 + 2–28 + 5 weeks), conventionally treated counterparts (N = 12, 23 + 5–30 weeks), and gestational age-matched controls (N = 8, 22 + 4–31 + 6 weeks). Immunoblotting, ELISA, and co-immunohistochemistry were used for biomarker analysis, including placental inflammation factors (iNOS, TNF-α). The results showed that TPE extended pregnancy by a median of 6.5 days in this cohort of early-onset preeclampsia. Serum sB7-H4, sFlt-1, and sEndoglin levels decreased, along with reduced expression of their membrane-bound proteins in placental tissue upon TPE treatment. Moreover, TPE-treated patients displayed reduced placental inflammation compared to preeclampsia patients receiving standard-of-care treatment. In conclusion, TPE may improve pregnancy outcomes in early-onset preeclampsia by lowering circulating levels of sB7-H4, sFlt-1, and sEndoglin, as well as reducing placental inflammation. This translational approach holds promise for enhancing placental function and extending gestation in high-risk pregnancies including very preterm PE or HELLP cases. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Pregnancy Complications)
Show Figures

Figure 1

Figure 1
<p>Serum levels of key biomarkers which decreased following therapeutic plasma exchange in early-onset PE patients. Serum levels of sFlt-1 (<b>A</b>), sFlt-1/PlGF (<b>B</b>), sEng (<b>C</b>), and sB7-H4 (<b>D</b>) before and after all the TPE treatment in PE patients. Data represent means ± SD. Significance: * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Serum levels of sFlt-1, sFlt-1/PlGF, and sEng, showing a decreased trend but not statistically significant before and after the first therapeutic plasma exchange in PE patients. Serum levels of sFlt-1 (<b>A</b>), sFlt-1/PlGF (<b>B</b>), sEng (<b>C</b>), and sB7-H4 (<b>D</b>) before and after the first TPE treatment in PE patients. Data represent means ± SD.</p>
Full article ">Figure 3
<p>Placental Flt-1, Eng, and B7-H4 protein levels were notably reduced in PE patients treated with TPE compared to those receiving the standard of care treatment. (<b>A</b>) Representative immunoblot images of Flt-1, Eng, and B7-H4 protein expression in the placenta of control (N = 8), PE (N = 12), and PE + TPE (N = 12) patients. The original blots are presented in <a href="#app1-ijms-25-11082" class="html-app">Supplementary Figure S1</a>. (<b>B</b>) Statistical analysis of A to represent the Flt-1 protein levels in placenta. (<b>C</b>) Statistical analysis of A to represent the Eng protein levels in placenta. (<b>D</b>) Statistical analysis of A to represent the B7-H4 protein levels in placenta. Data represent medians ± interquartile ranges with minimum/maximum values as whiskers. 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; 0.01.</p>
Full article ">Figure 4
<p>Placental Flt-1, Eng, and B7-H4 protein levels seem to be decreased in placental villi in PE patients treated with TPE compared to those receiving the standard-of-care treatment. (<b>A</b>–<b>C</b>) Double immunolabeling of Flt-1 (green) and CK7 (red) in placentas from controls and PE patients without and with TPE treatment. The STBs show strong Flt-1 staining in the untreated PE group. (<b>D</b>–<b>F</b>) Double immunolabeling of Eng (green) and CK7 (red), highlighting reduced Eng staining on the basal side of the STB in TPE-treated patients. (<b>G</b>–<b>I</b>) Double immunolabeling of B7-H4 (green) and CK7 (red), with the untreated PE group showing higher B7-H4 expression in the STB. DAPI was used to counterstain DNA (blue). The triangles point to the STB. The scale bar represents 75 μm.</p>
Full article ">Figure 5
<p>Placental inflammation was notably reduced in PE patients treated with TPE compared to those receiving the standard-of-care treatment. (<b>A</b>) Representative immunoblot images of TNF-α and iNOS protein expression in the placenta of control (N = 8), PE (N = 12), and PE + TPE (N = 12) patients. (<b>B</b>,<b>C</b>) Statistical analysis of A. (<b>D</b>) Double immunolabeling of iNOS (green) and CK7 (red) in placentas of control, PE, and PE + TPE patients. DAPI was used to counterstain DNA (blue). Data represent medians ± interquartile ranges with minimum/maximum values as whiskers. Significance: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. The scale bar represents 75 μm.</p>
Full article ">Figure 6
<p>Schematic diagram of TPE treatment in early-onset PE. TPE demonstrated efficacy in extending the gestational period for patients with early-onset PE. TPE removes a portion of maternal plasma, replacing it with fresh frozen plasma and/or 4% human albumin, leading to a reduction in these circulating factors. During plasma exchange, there is a notable reduction in the circulating maternal levels of sFlt-1, sEng, and sB7-H4, alongside a corresponding decrease in the membrane-bound proteins (Flt-1, Eng, and B7-H4) within placental tissues, which are the original source of these secreted proteins. This reduction aligns with an amelioration in placental functionality. Specifically, the observed decrease in placental inflammation (iNOS and TNF-α) and cellular apoptosis (cleaved caspase 3), along with the increase in proliferation (Cyclin D1, p21) after TPE treatment, may potentially be associated with the role of B7-H4. Diagram created with BioRender.com.</p>
Full article ">
12 pages, 2354 KiB  
Article
Vitamin D Receptor—Interplay in COVID-19-Negative, -Infected, and -Vaccinated Women during Pregnancy
by Constantin Condac, Ludmila Lozneanu, Daniela Roxana Matasariu, Alexandra Ursache, Iuliana Elena Bujor, Maria Elena Niță, Vasile Lucian Boiculese, Mihai Sava, Paula Țăroi and Victoria Bîrluțiu
J. Clin. Med. 2024, 13(20), 6140; https://doi.org/10.3390/jcm13206140 (registering DOI) - 15 Oct 2024
Viewed by 338
Abstract
Background: The trophoblast is a significant source of vitamin D synthesis during pregnancy, with the literature suggesting its role in fetal growth. We aim to underline a possible mechanism that would explain negative fetal outcomes in COVID-19-positive mothers by examining the relationship between [...] Read more.
Background: The trophoblast is a significant source of vitamin D synthesis during pregnancy, with the literature suggesting its role in fetal growth. We aim to underline a possible mechanism that would explain negative fetal outcomes in COVID-19-positive mothers by examining the relationship between altered placental structure and function and throphoblast cells‘ vitamin D receptor levels. Methods: The study included 170 placental samples collected from women who gave birth at term without complications, divided into three groups: COVID-19-positive and unvaccinated, COVID-19-negative and vaccinated, and COVID-19-negative and unvaccinated, with exclusion criteria for any other medical complications. Immunohistochemistry (IHC) was performed to detect vitamin D receptor (VDR) expression, and immediate fetal outcomes (weight and Apgar score) were assessed. Results: We found lower gestational age at birth, lower birth weight, and reduced placental VDR (vitamin D receptor) levels in COVID-19-positive women compared to COVID-19-vaccinated and COVID-19-negative women. Conclusions: The presence of the vitamin D receptor in the placenta is related to fetal and placental growth. Its deficiency may contribute to negative fetal outcomes in COVID-19-positive cases. Full article
(This article belongs to the Special Issue New Advances in COVID-19 and Pregnancy)
Show Figures

Figure 1

Figure 1
<p>Representative histopathological changes in placenta (H&amp;E). (<b>A</b>) COVID-19-positive pregnant women: small, well-vascularized chorionic villi. Syncytial knots and intervillous fibrin (HE × 10). (<b>B</b>) COVID-19-vaccinated pregnant women: chorionic villi, congestion, and fibrosis (HE × 20). (<b>C</b>) COVID-19-negative and unvaccinated pregnant women: different sizes of chorionic villi, congestion, and area of fibrosis (HE × 10).</p>
Full article ">Figure 2
<p>Representative images—immunohistochemical visualization of VDR in mononuclear cells of trophoblasts. (<b>A</b>) COVID-19-positive pregnant women (×20). (<b>B</b>) COVID-19-vaccinated pregnant women (×20). (<b>C</b>) COVID-19-negative and unvaccinated pregnant women (×10).</p>
Full article ">
18 pages, 30188 KiB  
Article
Intestinal Region-Dependent Impact of NFκB-Nrf Crosstalk in Myenteric Neurons and Adjacent Muscle Cells in Type 1 Diabetic Rats
by Bence Pál Barta, Benita Onhausz, Abigél Egyed-Kolumbán, Afnan AL Doghmi, János Balázs, Zita Szalai, Ágnes Ferencz, Edit Hermesz, Mária Bagyánszki and Nikolett Bódi
Biomedicines 2024, 12(10), 2347; https://doi.org/10.3390/biomedicines12102347 (registering DOI) - 15 Oct 2024
Viewed by 348
Abstract
Background/Objectives: Type 1 diabetes affects cytokines as potential inducers of NFκB signalling involved in inflammation and neuronal survival. Our goal was to assess the expression of NFκB p65 and its negative regulator, Nrf2, in myenteric neurons and adjacent smooth muscle of different gut [...] Read more.
Background/Objectives: Type 1 diabetes affects cytokines as potential inducers of NFκB signalling involved in inflammation and neuronal survival. Our goal was to assess the expression of NFκB p65 and its negative regulator, Nrf2, in myenteric neurons and adjacent smooth muscle of different gut segments after chronic hyperglycaemia and immediate insulin treatment. Methods: After ten weeks of hyperglycaemia, intestinal samples of control, streptozotocin-induced diabetic and insulin-treated diabetic rats were prepared for fluorescent immunohistochemistry, immunogold electron microscopy, ELISA and qPCR. Results: In the diabetic rats, the proportion of NFκB p65-immunoreactive myenteric neurons decreased significantly in the duodenum and increased in the ileum. The density of NFκB p65-labelling gold particles increased in the ileal but remained unchanged in the duodenal ganglia. Meanwhile, both total and nuclear Nrf2 density increased in the myenteric neurons of the diabetic duodenum. In smooth muscle, NFκB p65 and Nrf2 density increased in the small intestine of diabetic rats. While on the mRNA level, NFκB p65 and Nrf2 were induced, on the protein level, NFκB p65 increased and Nrf2 decreased in muscle/myenteric plexus homogenates. Insulin treatment had protective effects. Conclusions: Our findings reveal a segment-specific NFκB and Nrf expression in myenteric neurons and ganglionic muscular environments, which may contribute to regional neuronal survival and motility disturbances in diabetes. Full article
(This article belongs to the Special Issue Inflammation and Peripheral Nervous System)
Show Figures

Figure 1

Figure 1
<p>Relative levels of NFκB p65 mRNA. (<b>a</b>) Relative levels of NFκB p65 mRNA in tissue homogenates from different gut segments of control rats. The expression level of NFκB p65 mRNA was nearly three times higher in the colon than in the small intestinal segments of controls. Data were expressed as means ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 (compare with CD); <sup>oo</sup> <span class="html-italic">p</span> &lt; 0.01 (between CI and CC). CD—control duodenum, CI—control ileum, CC—control colon. (<b>b</b>) Effects of long-lasting hyperglycaemia and insulin treatment on the relative level of NFκB p65 mRNA in tissue homogenates from different gut segments. In the diabetics, the relative level of NFκB p65 mRNA displayed a more than 3-fold increase in the tissue homogenates of the colon and ileum, which was prevented by insulin treatment. Data were expressed as means ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 (compare to C); <sup>oo</sup> <span class="html-italic">p</span> &lt; 0.01 (between D and ID). C—controls, D—diabetics, ID—insulin-treated diabetics.</p>
Full article ">Figure 2
<p>Tissue levels of NFκB p65. (<b>a</b>) Tissue levels of NFκB p65 in intestinal smooth muscle/myenteric plexus homogenates from different gut segments of control rats. The tissue level of NFκB p65 displayed a significant decrease from the duodenum to the colon of controls. Data were expressed as means ± SEM. * <span class="html-italic">p</span> &lt; 0.05 (compare with CD). CD—control duodenum, CI—control ileum, CC—control colon. (<b>b</b>) Effects of long-lasting hyperglycaemia and insulin treatment on the tissue levels of NFκB p65 in smooth muscle/myenteric plexus homogenates from different gut segments. In the diabetic rats, the NFκB p65 level was doubled in the duodenum and tripled in the ileum, while it did not change in the colon. Immediate insulin treatment was completely protective against diabetes-related changes. Data were expressed as means ± SEM. * <span class="html-italic">p</span> &lt; 0.05 (compare with C); <sup>o</sup> <span class="html-italic">p</span> &lt; 0.05 (between D and ID). C—controls, D—diabetics, ID—insulin-treated diabetics.</p>
Full article ">Figure 3
<p>Representative fluorescent micrographs of whole-mount preparations of myenteric ganglia from the duodenum and ileum of control, diabetic and insulin-treated diabetic rats after NFκB p65-HuC/HuD double-labelling immunohistochemistry. HuC/HuD as a pan-neuronal marker was applied to label myenteric neurons. CD—control duodenum, CI—control ileum, DD—diabetic duodenum, DI—diabetic ileum, IDD—insulin-treated diabetic duodenum, IDI—insulin-treated diabetic ileum, arrows—NFκB p65-immunoreactive myenteric neurons. Scale bars: 20 μm.</p>
Full article ">Figure 3 Cont.
<p>Representative fluorescent micrographs of whole-mount preparations of myenteric ganglia from the duodenum and ileum of control, diabetic and insulin-treated diabetic rats after NFκB p65-HuC/HuD double-labelling immunohistochemistry. HuC/HuD as a pan-neuronal marker was applied to label myenteric neurons. CD—control duodenum, CI—control ileum, DD—diabetic duodenum, DI—diabetic ileum, IDD—insulin-treated diabetic duodenum, IDI—insulin-treated diabetic ileum, arrows—NFκB p65-immunoreactive myenteric neurons. Scale bars: 20 μm.</p>
Full article ">Figure 4
<p>Proportion of NFκB p65-immunoreactive myenteric neurons in the duodenum and ileum of control, diabetic and insulin-treated diabetic rats. In the diabetics, the proportion of NFκB p65-immunoreactive myenteric neurons was significantly decreased in the duodenum and increased in the ileum, which was prevented by immediate insulin treatment. Data were expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (compare with C); <sup>o</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>oooo</sup> <span class="html-italic">p</span> &lt; 0.0001 (between D and ID). C—controls, D—diabetics, ID—insulin-treated diabetics.</p>
Full article ">Figure 5
<p>Representative electron micrographs of portions of the perikaryon and nuclei of myenteric neurons from ileum and intestinal smooth muscle cells from duodenum of control, diabetic and insulin-treated diabetic rats after NFκB p65 post-embedding immunohistochemistry. CD—control duodenum, CI—control ileum, DD—diabetic duodenum, DI—diabetic ileum, IDD—insulin-treated diabetic duodenum, IDI—insulin-treated diabetic ileum, arrows—18 nm gold particles’ labelling NFκB p65. Scale bars: 250 nm.</p>
Full article ">Figure 6
<p>Quantification of gold particles’ labelling NFκB p65 in myenteric ganglia (<b>a</b>) and intestinal smooth muscle (<b>b</b>) from different gut segments of control, diabetic and insulin-treated diabetic rats. The number of NFκB p65-labelling gold particles increased in the ileal myenteric ganglia and intestinal smooth muscle of both the duodenum and the ileum of diabetic animals relative to the controls, which was prevented by insulin. Data were expressed as means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (compare with C); <sup>o</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>oo</sup> <span class="html-italic">p</span> &lt; 0.01 (between D and ID). C—controls, D—diabetics, ID—insulin-treated diabetics.</p>
Full article ">Figure 7
<p>Relative levels of Nrf2 mRNA. (<b>a</b>) Relative levels of Nrf2 mRNA in tissue homogenates from different gut segments of control rats. The expression level of Nrf2 mRNA was multiple times higher in the colon than in the small intestinal segments of the controls. Data were expressed as means ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 (compare with CD); <sup>oooo</sup> <span class="html-italic">p</span> &lt; 0.0001 (between CI and CC). CD—control duodenum, CI—control ileum, CC—control colon. (<b>b</b>) Effects of long-lasting hyperglycaemia and insulin treatment on the relative level of Nrf2 mRNA in tissue homogenates from different gut segments. In the diabetics, the relative level of Nrf2 mRNA displayed a robust increase in all segments along the duodenum–ileum–colon axis, which was prevented by insulin treatment. Data were expressed as means ± 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 (compare with C); <sup>oo</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>ooo</sup> <span class="html-italic">p</span> &lt; 0.001 (between D and ID). C—controls, D—diabetics, ID—insulin-treated diabetics.</p>
Full article ">Figure 8
<p>Tissue levels of Nrf2. (<b>a</b>) Tissue levels of Nrf2 in intestinal smooth muscle/myenteric plexus homogenates from different gut segments of control rats. The tissue levels of Nrf2 were significantly higher in the small intestine than the colon of the controls. Data were expressed as means ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 (compare with CD); <sup>o</sup> <span class="html-italic">p</span> &lt; 0.05 (between CI and CC). CD—control duodenum, CI—control ileum, CC—control colon. (<b>b</b>) Effects of long-lasting hyperglycaemia and insulin treatment on the tissue levels of Nrf2 in smooth muscle/myenteric plexus homogenates from different gut segments. In the diabetic rats, the Nrf2 level was decreased in the duodenum and ileum, while it did not change in the colon. Immediate insulin treatment was protective against diabetes-related changes. Data were expressed as means ± SEM. ** <span class="html-italic">p</span> &lt; 0.01 (compare with C); <sup>o</sup> <span class="html-italic">p</span> &lt; 0.05 (between D and ID). C—controls, D—diabetics, ID—insulin-treated diabetics.</p>
Full article ">Figure 9
<p>Representative fluorescent micrograph of a whole-mount preparation of myenteric ganglia from the ileum of a control rat after NFκB p65-Nrf2-Peripherin triple-labelling immunohistochemistry (<b>a</b>). Pan-neuronal Peripherin was used to label myenteric neurons. Representative electron micrograph of a portion of the perikaryon and nucleus of a myenteric neuron from duodenum of a control rat after NFκB p65-Nrf2 post-embedding immunohistochemistry (<b>b</b>). Red circles—10 nm gold particles’ labelling NFκB p65, arrows—18 nm gold particles’ labelling Nrf2. CI—control ileum, CD—control duodenum. Scale bars: 20 µm (<b>a</b>), 250 nm (<b>b</b>).</p>
Full article ">Figure 10
<p>Representative electron micrographs of portions of the perikaryon and nuclei of myenteric neurons (<b>a</b>) and intestinal smooth muscle cells (<b>b</b>) from the duodenum of control and diabetic rats after Nrf2 post-embedding immunohistochemistry. CD—control duodenum, DD—diabetic duodenum. Arrows—18 nm gold particles’ labelling Nrf2 in myenteric neurons, arrowheads—18 nm gold particles’ labelling Nrf2 in muscle cells. Scale bars: 250 nm.</p>
Full article ">Figure 11
<p>Quantification of gold particles’ labelling Nrf2 in myenteric ganglia (<b>a</b>) and intestinal smooth muscle (<b>b</b>) from the duodenum of control, diabetic and insulin-treated diabetic rats. The number of Nrf2-labelling gold particles increased in the myenteric ganglia and smooth muscle of diabetic duodenum relative to controls, which was partially prevented by insulin treatment. Data were expressed as means ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (compare with C); <sup>oo</sup> <span class="html-italic">p</span> &lt; 0.01 (between D and ID). C—controls, D—diabetics, ID—insulin-treated diabetics.</p>
Full article ">
14 pages, 1964 KiB  
Article
High Ki-67 Expression Predicting a Risk Factor for the Progression of Disease within 24 Months and Microenvironment in Follicular Lymphoma
by Hinako Narita, Kai Kuroiwa, Yukiko Kawaguchi, So Murai, Yosuke Sasaki, Mayumi Homma, Natsuki Kawamata, Hidenori Hayashi, Kazuki Nagao, Reiko Okamura, Yuka Uesugi, Yohei Sasaki, Shotaro Shimada, Megumi Watanuki, Nana Arai, Kouji Yanagisawa, Eisuke Shiozawa, Toshiko Yamochi and Norimichi Hattori
Int. J. Mol. Sci. 2024, 25(20), 11057; https://doi.org/10.3390/ijms252011057 (registering DOI) - 15 Oct 2024
Viewed by 228
Abstract
Most follicular lymphomas (FLs) demonstrate an indolent clinical course with favorable outcomes; however, a fraction of patients experiences progression of disease within 24 months (POD24) and has adverse outcomes. This study aimed to determine the predictive risk factors for POD24 in patients with [...] Read more.
Most follicular lymphomas (FLs) demonstrate an indolent clinical course with favorable outcomes; however, a fraction of patients experiences progression of disease within 24 months (POD24) and has adverse outcomes. This study aimed to determine the predictive risk factors for POD24 in patients with FL, and the characteristics of the microenvironment in FL with POD24. By multivariate analysis, we revealed that increased Ki-67 expression was associated with POD24 events in patients with FL (hazard ratio [HR]: 6.29, 95% confidence interval [CI]: 1.96–20.22, p = 0.0020). Additionally, patients with FL with POD24 demonstrated immune cell reduction by immunohistochemistry analysis. Our results help better understand the therapeutic strategies for FL with POD24. Full article
Show Figures

Figure 1

Figure 1
<p>Overall survival in patients with FL with and without POD24.</p>
Full article ">Figure 2
<p>Comparison of immune cells between patients with FL with and without POD24 according to separately counted intrafollicular and interfollicular areas. (<b>a</b>) CD3, (<b>b</b>) CD4, (<b>c</b>) CD8, and (<b>d</b>) CD68. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Immunohistochemical staining (magnification, 100×) of the intrafollicular area (black asterisks) and interfollicular area (black triangles) in samples of patients with FL and POD24 (66 years, male) and non-POD24 (63 years, female). Staining for (<b>a</b>) CD3, (<b>b</b>) CD4, (<b>c</b>) CD8, and (<b>d</b>) CD68.</p>
Full article ">
8 pages, 3561 KiB  
Communication
Association of Senescence Markers with Age and Allograft Rejection in Renal Transplant Recipients
by Peter Vavrinec, Jakub Krivy, Sona Sykorova, Helena Bandzuchova, Zuzana Zilinska and Diana Vavrincova-Yaghi
Biomedicines 2024, 12(10), 2338; https://doi.org/10.3390/biomedicines12102338 - 14 Oct 2024
Viewed by 314
Abstract
Background/Objectives: Renal transplantation is the treatment of choice for patients with end-stage renal disease. In the last decade, the number of older renal transplant recipients has significantly increased. However, these patients are at a higher risk of developing post-transplant complications. Therefore, identifying the [...] Read more.
Background/Objectives: Renal transplantation is the treatment of choice for patients with end-stage renal disease. In the last decade, the number of older renal transplant recipients has significantly increased. However, these patients are at a higher risk of developing post-transplant complications. Therefore, identifying the suitable biomarkers to predict which older patients are at risk of complications is crucial. Cellular senescence could provide insights into the increased vulnerability in this population and guide personalized post-transplant care. Methods: This preliminary study involved biopsies from 25 patients with renal allograft rejection and 18 patients without rejection, further divided into older (50–65 years) and younger (29–40 years) groups. Biopsies were collected at different time points after transplantation, and rejection was classified according to the histological Banff 07 criteria. Additionally, immunohistochemistry for the markers of cellular senescence, p27kip1 and p16INK4a, was performed. Results: We observed that the number of p27kip1-positive glomeruli was higher in the older patients with rejection compared to the younger patients with rejection, and a similar pattern was found in the patients without rejection. However, the number of p27kip1-positive tubules was higher in the older patients with rejection compared to the younger patients with rejection, as well as compared to both the older and younger patients without rejection. Tubular p16INK4a expression was not significantly different in the older patients with rejection compared to the younger patients with rejection, and the same pattern was observed in the patients without rejection. However, it was increased in the older patients with rejection in comparison to the older patients without rejection. Conclusions: Our preliminary data suggest the strong potential of both p16INK4a and p27kip1 as biomarkers of renal graft rejection, particularly in older renal transplant recipients. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

Figure 1
<p>Expression of p27<sup>kip1</sup> (glomeruli and tubules) and p16<sup>INK4a</sup> (tubules) in older and younger patients with allograft rejection after kidney transplantation. Localization of p27<sup>kip1</sup> and p16<sup>INK4a</sup> proteins in the graft biopsies. (<b>A</b>) Glomerular p27<sup>kip1</sup> expression was increased in older patients compared to younger patients in the group w/rj, as well as in older patients compared to younger patients in the w/o rj group; (<b>B</b>) Tubular p27<sup>kip1</sup> expression was increased in older patients w/rj compared to younger patients w/rj, as well as to both older (<span class="html-italic">p</span> = 0.06) and younger patients w/o rj; (<b>C</b>) Tubular p16<sup>INKa</sup> expression was increased in older patients w/rj compared to older patients w/o rj; (<b>D</b>) p27<sup>kip1</sup> expression in the glomeruli of older patients and (<b>E</b>) younger patients w/rj and (<b>F</b>) older patients and (<b>G</b>) younger patients w/o rj. (<b>H</b>) Tubular p27<sup>kip1</sup> expression in older patients and (<b>I</b>) younger patients w/rj and (<b>J</b>) older patients and (<b>K</b>) younger patients w/o rj. (<b>L</b>) Tubular p16<sup>INK4a</sup> expression in older patients and (<b>M</b>) younger patients w/rj and (<b>N</b>) older patients and (<b>O</b>) younger patients w/o rj. Magnification (400×). * <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; w/rj, with rejection; w/o rj, without rj. Data are presented as mean ± SD.</p>
Full article ">Figure 1 Cont.
<p>Expression of p27<sup>kip1</sup> (glomeruli and tubules) and p16<sup>INK4a</sup> (tubules) in older and younger patients with allograft rejection after kidney transplantation. Localization of p27<sup>kip1</sup> and p16<sup>INK4a</sup> proteins in the graft biopsies. (<b>A</b>) Glomerular p27<sup>kip1</sup> expression was increased in older patients compared to younger patients in the group w/rj, as well as in older patients compared to younger patients in the w/o rj group; (<b>B</b>) Tubular p27<sup>kip1</sup> expression was increased in older patients w/rj compared to younger patients w/rj, as well as to both older (<span class="html-italic">p</span> = 0.06) and younger patients w/o rj; (<b>C</b>) Tubular p16<sup>INKa</sup> expression was increased in older patients w/rj compared to older patients w/o rj; (<b>D</b>) p27<sup>kip1</sup> expression in the glomeruli of older patients and (<b>E</b>) younger patients w/rj and (<b>F</b>) older patients and (<b>G</b>) younger patients w/o rj. (<b>H</b>) Tubular p27<sup>kip1</sup> expression in older patients and (<b>I</b>) younger patients w/rj and (<b>J</b>) older patients and (<b>K</b>) younger patients w/o rj. (<b>L</b>) Tubular p16<sup>INK4a</sup> expression in older patients and (<b>M</b>) younger patients w/rj and (<b>N</b>) older patients and (<b>O</b>) younger patients w/o rj. Magnification (400×). * <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; w/rj, with rejection; w/o rj, without rj. Data are presented as mean ± SD.</p>
Full article ">
19 pages, 5218 KiB  
Article
Chinese Herbal Medicines as Natural Alternative Products to Antibiotics in Weaned Piglets through Intestinal Microbiota Regulation
by Che-Hsuan Wang, Kou-Toung Chung, Li-Yu Su, Wan-Jhen Wu, Pei-Hwa Wang, Ming-Chung Lee, Szu-Chuan Shen and Chung-Hsin Wu
Int. J. Mol. Sci. 2024, 25(20), 11034; https://doi.org/10.3390/ijms252011034 - 14 Oct 2024
Viewed by 421
Abstract
During the growth process of weaned piglets, digestive problems such as gastrointestinal disorders and diarrhea are common. Farmers usually use antibiotics to help piglets grow smoothly. However, the overuse of antibiotics can lead to antibiotic resistance issues. Therefore, this study chose to use [...] Read more.
During the growth process of weaned piglets, digestive problems such as gastrointestinal disorders and diarrhea are common. Farmers usually use antibiotics to help piglets grow smoothly. However, the overuse of antibiotics can lead to antibiotic resistance issues. Therefore, this study chose to use plant extracts as feed additives to explore their potential as alternatives to antibiotics. Additionally, Tilmicosin was used as the antibiotic because it is widely used in treating respiratory infections in piglets. Since traditional Chinese medicine often uses natural products, we selected Guizhi Li-Zhong (GLZ) extract as an alternative to antibiotics. The experiment involved 126 piglets, each 4 weeks old, which were randomly assigned to one of four groups: the sham group (basal diet without supplements, 10.3 ± 0.4 kg, n = 31), the low-dose GLZ group (basal diet with 0.05% GLZ, 10.9 ± 0.4 kg, n = 32), the regular-dose GLZ group (basal diet with 0.2% GLZ, 10.6 ± 0.4 kg, n = 32), and the regular-dose Tilmicosin antibiotic group (basal diet with 0.2% Tilmicosin, 10.2 ± 0.3 kg, n = 31). We recorded and compared the survival rate, growth rate, feed conversion ratio, and diarrhea incidence among four groups of weaned piglets from the 4th to the 10th weeks of age. Then, we examined the oxidative stress, inflammation, and apoptosis in small intestine tissue (jejunum and ileum) through immunohistochemistry and Western blot and compared the gut microbiota in large intestine tissue (colon and rectum) through a next-generation sequencing (NGS) analysis. Our results showed that weaned piglets supplemented with 0.05% and 0.2% GLZ had better survival rates, growth rates (p < 0.01), and feed conversion ratios (p < 0.01) compared to those receiving sham treatment. Even weaned piglets supplemented with 0.2% GLZ performed better than those supplemented with 0.2% Tilmicosin antibiotics (p < 0.05). Furthermore, the incidence of diarrhea and small intestine injury (indicated by oxidative stress-, inflammation-, and apoptosis-related proteins) in piglets supplemented with 0.05% and 0.2% GLZ was lower than in piglets receiving sham treatment (p < 0.05). Even piglets supplemented with 0.2% GLZ had less injury than those supplemented with 0.2% Tilmicosin antibiotics (p < 0.05). The NGS results further showed that GLZ treatment significantly improved beneficial bacteria in weaned piglets (p < 0.05), while antibiotic treatment reduced beneficial bacteria (p < 0.05). In summary, we recommend adding GLZ to the feed as an alternative to antibiotics. This not only effectively reduces intestinal damage but also improves the gut microbiota, thereby promoting the growth of weaning piglets. Full article
(This article belongs to the Special Issue Drug Discovery and Development Focusing on Natural Products)
Show Figures

Figure 1

Figure 1
<p>Chromatographic fingerprints of the dietary herbal formula of GLZ. Bioactive marker substances of GLZ are listed on the upper right and were qualitatively determined within 65 min by 3D HPLC. AU, arbitrary perfusion unit.</p>
Full article ">Figure 2
<p>Antioxidant capacity and cytotoxicity of GLZ treatment. (<b>A</b>) DPPH free radical method under 1–500 μg/mL GLZ treatments. Dotted line indicates 75% of free radical scavenging activity. (<b>B</b>) Quantified Caco-2 cell viability under 1–500 μg/mL GLZ treatments by MTT assay. Dotted line indicates 100% of Caco-2 cell viability. Data are shown as the mean ± SEM, and the sample number must be at least 3 times for each GLZ treatment.</p>
Full article ">Figure 3
<p>Comparison of tissue morphology of jejunum and ileum among four groups of weaned piglets. (<b>A</b>) Representative H&amp;E stains of jejunum and ileum tissue among four groups of weaned piglets. Scale bars = 300 μm. (<b>B</b>) Quantified length of the intestinal villi (jejunum and ileum) among four groups of weaned piglets (N = 3 for each group). ** indicates differences <span class="html-italic">p</span> &lt; 0.01 between intestinal villi. SH group, sham treatment; LD group, low-dose 0.05% GLZ addition; RD group, regular-dose 0.2% GLZ addition; and RT group, regular-dose antibiotic 0.2% Tilmicosin addition.</p>
Full article ">Figure 4
<p>Comparison of antioxidant-related SOD2 expressions in the small intestine tissue among four groups of weaned piglets. (<b>A</b>) Representative IHC stains of SOD2 expressions in jejunum and ileum tissue among weaned piglets with SH, LD, RD, and RT treatments. SOD2 expressions are marked with dark brown color. Scale bars = 300 μm. (<b>B</b>) Western blotting expressions of SOD2 in the small intestine tissue among weaned piglets with SH, LD, RD, and RT treatments. Right bar chart shows quantified SOD2 expressions relative to β-actin in the small intestine tissue among four groups of weaned piglets (<span class="html-italic">n</span> = 3 for each group). SH group, sham treatment; LD group, low-dose 0.05% GLZ addition; RD group, regular-dose 0.2% GLZ addition; and RT group, regular-dose antibiotic 0.2% Tilmicosin addition. Data are shown as the mean ± SEM (** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 compared with the SH group; one-way ANOVA followed by Student–Newman–Keuls multiple comparison posttest).</p>
Full article ">Figure 5
<p>Comparison of inflammation-related TNF-α expressions in the small intestine tissue among four groups of weaned piglets. (<b>A</b>) Representative IHC stains of TNF-α expressions in jejunum and ileum tissue among weaned piglets with SH, LD, RD, and RT treatments. TNF-α expressions are marked with dark brown color. Scale bars = 300 μm. (<b>B</b>) Western blotting expressions of TNF-α in the small intestine tissue among weaned piglets with SH, LD, RD, and RT treatments. Right bar chart shows quantified TNF-α expressions relative to β-actin in the small intestine tissue among four groups of weaned piglets (<span class="html-italic">n</span> = 3 for each group). SH group, sham treatment; LD group, low-dose 0.05% GLZ addition; RD group, regular-dose 0.2% GLZ addition; and RT group, regular-dose antibiotic 0.2% Tilmicosin addition. (** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 compared with the SH group; one-way ANOVA followed by Student–Newman–Keuls multiple comparison posttest).</p>
Full article ">Figure 6
<p>Comparison of apoptosis-related caspase 3 expressions in the small intestine tissue among four groups of weaned piglets. (<b>A</b>) Representative IHC stains of caspase 3 expressions in jejunum and ileum tissue among weaned piglets with SH, LD, RD, and RT treatments. Caspase 3 expressions are marked with dark brown color. Scale bars = 300 μm. (<b>B</b>) Western blotting expressions of caspase 3 in the small intestine tissue among weaned piglets with SH, LD, RD, and RT treatments. Right bar chart shows quantified caspase 3 expressions relative to β-actin in the small intestine tissue among four groups of weaned piglets (<span class="html-italic">n</span> = 3 for each group). SH group, sham treatment; LD group, low-dose 0.05% GLZ addition; RD group, regular-dose 0.2% GLZ addition; and RT group, regular-dose antibiotic 0.2% Tilmicosin addition. (** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 compared with the SH group; one-way ANOVA followed by Student–Newman–Keuls multiple comparison posttest).</p>
Full article ">Figure 7
<p>Gut microbial community structure in the colon and rectum of the large intestine tissue among four groups of weaned piglets. The microbial community bar plot in the colon and rectum tissue of weaned piglets in (<b>A</b>) phylum level (a) relative abundance of most abundant OTUs in phyla level of each sample, and (b) quantified relative abundance of Bacteroidetes, Firmicutes, Fusobacteria, and Proteobacteria in phyla level of each sample; (<b>B</b>) Class level (a) relative abundance of most abundant OTUs in class level of each sample, and (b) quantified relative abundance of Bacteroidia, Clostridia, and Spirochaetes in class level of each sample; (<b>C</b>) Order level (a) relative abundance of most abundant OTUs in order level of each sample, and (b) quantified relative abundance of Bacteroidales, Clostridiales, and Lactobacillales in order level of each sample; (<b>D</b>) Family level (a) relative abundance of most abundant OTUs in family level of each sample, and (b) quantified relative abundance of Lachnospiraceae, Lactobacillaceae, Prevotellaceae, and Ruminococcaceae, in family level of each sample; (<b>E</b>) Genus level (a) relative abundance of most abundant OTUs in genus level of each sample, and (b) quantified relative abundance of Anaerovibrio, Faecalibacterium, Prevotella, and Roseburia in genus level of each sample; (<b>F</b>) Species level (a) relative abundance of most abundant OTUs in species level of each sample, and (b) quantified relative abundance of <span class="html-italic">Lactobacillus_reuteri</span>, <span class="html-italic">Prevotella_copri</span>, <span class="html-italic">Prevotella_stercorea,</span> and <span class="html-italic">Roseburia_faecis</span>. SH group, sham treatment; LD group, low-dose 0.05% GLZ addition; RD group, regular-dose 0.2% GLZ addition; and RT group, regular-dose antibiotic 0.2% Tilmicosin addition. (<span class="html-italic">n</span> = 3 for each group, ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 compared with the SH group; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared with the colon group, two-way ANOVA followed by Student–Newman–Keuls multiple comparison posttest).</p>
Full article ">
10 pages, 1090 KiB  
Article
NTRK Gene Expression Analysis in Oral Squamous Cell Carcinoma Mexican Population
by Lilibeth Stephania Escoto-Vasquez, Javier Portilla-Robertson, Josué Orlando Ramírez-Jarquín, Luis Fernando Jacinto-Alemán, Alejandro Alonso-Moctezuma, Carla Monserrat Ramírez-Martínez, Osmar Alejandro Chanes-Cuevas and Fabiola Salgado-Chavarria
Dent. J. 2024, 12(10), 327; https://doi.org/10.3390/dj12100327 - 14 Oct 2024
Viewed by 422
Abstract
Oral cancer holds the sixth position in malignancies worldwide; 90% correspond to oral squamous cell carcinoma (OSCC). Diverse reports suggest that NTRK genes and their receptors are key oncogenesis regulators to tumor progression in human cancers. Objective: To analyze the NTRK and Trk [...] Read more.
Oral cancer holds the sixth position in malignancies worldwide; 90% correspond to oral squamous cell carcinoma (OSCC). Diverse reports suggest that NTRK genes and their receptors are key oncogenesis regulators to tumor progression in human cancers. Objective: To analyze the NTRK and Trk expression and their association with clinicopathological features of OSCC in Mexican patients’ samples. Material and Methods: We analyzed 95 OSCC cases of pan-trk immunoexpression through a software-assisted method. Gene expression was analyzed by RT-qPCR employing the ΔΔCT method. Kruskal–Wallis and Spearman’s correlation tests were performed. Results: Our mean age was 62.4 (±16.9) years. A total of 37 cases were tumors in the lateral border of the tongue. Age was significantly associated with the anatomical site. 42% (40 of 95) cases were pan-trk positive. A total of 21 cases showed intense immunoexpression predominantly in poorly differentiated OSCC, with a significant correlation between immunoexpression and age and gender. Gene expression showed that poorly differentiated cases exhibited higher NTRK2 expression, while well-differentiated cases demonstrated NTRK3 significantly higher expression. Conclusions: Our results suggest that NTRK family expression is present in OSCC, with differential expression related to differentiation degree. Additional information about their activation or mutational status could reinforce their potential as a possible primary or adjuvant treatment target. Full article
(This article belongs to the Special Issue Molecular Diagnostics in Oral Diseases: Volume II)
Show Figures

Figure 1

Figure 1
<p>OSCC histological and immunohistochemical features. (<b>a</b>) Invasive islet of well-differentiated OSCC with cellular keratinization (asterisk), (<b>b</b>) OSCC moderately differentiated with cellular and nuclear pleomorphism (bracket), (<b>c</b>) poorly differentiated OSCC with the presence of fusiform and pleomorphic cells (bracket), (<b>d</b>) cytoplasmic mild pan-trk immunoexpression in well-differentiated OSCC (arrow), (<b>e</b>) perinuclear moderate pan-trk immunoexpression in moderate differentiated OSCC (arrow), and (<b>f</b>) strong perinuclear pan-trk immunoexpression in poorly differentiated OSCC (arrow). All photomicrographs were obtained at 400× magnification. Scale bar: 20 μm.</p>
Full article ">Figure 2
<p>NTRK1-3 ΔΔCT results according to their differentiation degree (WD = well differentiated, MD = moderately differentiated, and PD = poorly differentiated).</p>
Full article ">
16 pages, 1188 KiB  
Article
Comprehensive Analysis of Receptor Status, Histopathological Classifications (B1–B5), and Cumulative Histological Dimensions in Breast Cancer: Predictors of Malignancy and Diagnostic Implications
by Oana Maria Burciu, Ioan Sas, Adrian-Grigore Merce, Simona Cerbu, Aurica Elisabeta Moatar, Anca-Elena Eftenoiu and Ionut Marcel Cobec
Cancers 2024, 16(20), 3471; https://doi.org/10.3390/cancers16203471 - 14 Oct 2024
Viewed by 370
Abstract
Introduction: Breast cancer has become one of the most serious and widespread public health concerns globally, affecting an increasing number of women—and, in rare cases, men—across the world. It is the most common cancer among women across all countries. In this study, we [...] Read more.
Introduction: Breast cancer has become one of the most serious and widespread public health concerns globally, affecting an increasing number of women—and, in rare cases, men—across the world. It is the most common cancer among women across all countries. In this study, we aimed to evaluate the influence of demographic factors, medical and reproductive history, diagnostic techniques, and hormone receptor status on the development and progression of breast cancer. Materials and Methods: A total of 687 female patients from Romania underwent standard breast examination techniques, including clinical breast examination, mammography, ultrasonography, and, ultimately, breast biopsy. Statistical analysis was performed using the R programming language and RStudio software. The study included a comparative analysis and a prediction analysis for malignancy and tumor size (cumulative histological dimension) through logistic and linear regression models. Results: The comparative analysis identified several variables associated with malignancy: older age (p < 0.001), non-vulnerability (p = 0.04), no daily physical activity (p = 0.002), no re-biopsy (p < 0.001), immunohistochemistry use (p < 0.001), use of larger gauge needles (p < 0.001), ultrasound-guided biopsy (p < 0.001), and vacuum biopsy (p < 0.001). The hormone receptor statuses—estrogen receptor (ER), progesterone receptor (PR), and androgen receptor (AR)—showed statistically significant differences in distribution across breast cancer B classifications. Logistic regression analysis identified ER, PR, and age as significant predictors of malignancy. Linear regression analysis revealed histopathological results, living environment, geographical region, vulnerability, prior breast examination, and the number of histological fragments as significant predictors of cumulative histological dimension. Conclusions: Our predictive models demonstrate the impact of demographic factors, medical history, diagnostic techniques, and hormone receptor status on breast cancer development and progression, accounting for a significant portion of the variance in malignancy and cumulative histological dimension. Full article
Show Figures

Figure 1

Figure 1
<p>Distribution of Cases Across B Classification. X-axis—B Classification, Y-axis—the percentage of cases within each group.</p>
Full article ">Figure 2
<p>Comparison of Age Distribution Between Benign and Malignant Breast Lesions.</p>
Full article ">Figure 3
<p>Comparison Between Immunohistochemistry Use and Histopathological Outcome (Malignant vs. Benign). The y-axis represents the proportions of cases. Green bars indicate malignant cases, and orange bars indicate benign cases. The groups are classified as 1 for the immunohistochemistry group and 0 for the non-immunohistochemistry group.</p>
Full article ">
12 pages, 5398 KiB  
Article
Biomarker Profiles and Clinicopathological Features in Head and Neck Squamous Cell Carcinoma Patients
by Timea Szatmari, Simona Mocan, Cristian Mircea Neagos and Zsuzsanna Pap
Medicina 2024, 60(10), 1681; https://doi.org/10.3390/medicina60101681 - 14 Oct 2024
Viewed by 414
Abstract
Background and Objectives: Head and neck squamous cell carcinomas (HNSCCs) vary significantly in terms of invasiveness, growth rate, and metastatic potential. This study aimed to investigate the expression of several prognostic biomarkers (Ki67, p53, EGFR, COX-2, Cx43, and p16) in HNSCC from various [...] Read more.
Background and Objectives: Head and neck squamous cell carcinomas (HNSCCs) vary significantly in terms of invasiveness, growth rate, and metastatic potential. This study aimed to investigate the expression of several prognostic biomarkers (Ki67, p53, EGFR, COX-2, Cx43, and p16) in HNSCC from various anatomical regions and to correlate these expressions with clinicopathological parameters. Materials and Methods: We performed immunohistochemistry on 91 histologically verified HNSCC cases from the County Emergency Hospital, Targu Mures. Biomarker expression for Ki67, COX-2, and Cx43 was assessed using a standard immunoexpression scoring system: S1: 0–10%, S2: 11–25%, S3: 26–50%, S4 > 50%; EGFR was scored based on membrane staining intensity: 0, 1+, 2+, 3+; we classified p16 as positive or negative; p53 was grouped into mutant and wild-type; and we compared these across histopathological types, tumor grades, anatomical locations, gender, and different age groups. We performed a comparative analysis of Cx43 expression levels in relation to the expression of the rest of the markers. Statistical analysis was conducted using GraphPad InStat 3 software, version 3.06 (GraphPad Software Inc., San Diego, USA). Results: The majority of tumors were in males (95.6%) aged 51–60 years. Mutant p53 expression was prevalent in most cases. Elevated Ki67 and EGFR expression were associated with more aggressive tumors. COX-2 levels varied, with a higher proportion of moderate and high immunoexpression (S3 + S4) observed in patients under 70 years old. Cx43 expression was generally low, especially in extralaryngeal tumors. Conclusions: HNSCC primarily affects older males, with the larynx being the most common site. High levels of Ki-67 and EGFR suggest more aggressive tumors, while low COX-2 levels reflect varying prognoses. Women may develop more aggressive tumors, and extralaryngeal tumors often present with more challenging prognoses. Low Cx43 expression may be more likely to coincide with higher Ki67 and COX-2 levels, possibly indicating a link with more aggressive tumor behavior. Full article
Show Figures

Figure 1

Figure 1
<p>Immunohistochemical staining: (<b>A</b>): Ki 67 intense positive nuclear staining in less than 10% of tumor cells, ×4; (<b>B</b>): Ki 67: intense positive nuclear staining in more than 50% of tumor cells, ×4; (<b>C</b>): Cx43 membrane reaction of weak intensity in less than 10% of the tumor cells, ×4; (<b>D</b>): Cx43 membrane reaction of increase intensity in 20% of the tumor cells, ×4.</p>
Full article ">Figure 2
<p>Immunohistochemical staining. (<b>A</b>): p53 aberrant mutational reaction, positive type, with over 90% of the tumor cells presenting a strong nuclear reaction, ×10; (<b>B</b>): p53 aberrant mutational reaction, negative type, with no nuclear reaction in tumor cells, ×10; (<b>C</b>): p53 normal wild-type expression, with nuclear reaction with variable intensity in some of the tumor cells, ×10.</p>
Full article ">Figure 3
<p>Immunohistochemical staining: (<b>A</b>): EGFR score 2+ with moderate membranous staining for at least 10% of tumor cells, ×4; (<b>B</b>): EGFR score 3+ with almost 100% of tumor cells with strong membranous staining, ×4; (<b>C</b>): COX-2: cytoplasmic positive and intense expressions in more than 50% of squamous cell carcinoma, ×4; (<b>D</b>): COX-2: cytoplasmic immunohistochemical reaction with variable intensity, ×4.</p>
Full article ">Figure 4
<p>Cx43 expression in relation to p53 expression.</p>
Full article ">Figure 5
<p>The relationship between Cx43 and Ki67 expression.</p>
Full article ">Figure 6
<p>The relationship between Cx43 and COX-2 expression.</p>
Full article ">Figure 7
<p>Cx43 expression in association with EGFR expression.</p>
Full article ">
23 pages, 4534 KiB  
Article
Comprehensive Analysis of the 5xFAD Mouse Model of Alzheimer’s Disease Using dMRI, Immunohistochemistry, and Neuronal and Glial Functional Metabolic Mapping
by Emil W. Westi, Saba Molhemi, Caroline Termøhlen Hansen, Christian Stald Skoven, Rasmus West Knopper, Dashne Amein Ahmad, Maja B. Rindshøj, Aishat O. Ameen, Brian Hansen, Kristi A. Kohlmeier and Blanca I. Aldana
Biomolecules 2024, 14(10), 1294; https://doi.org/10.3390/biom14101294 - 13 Oct 2024
Viewed by 765
Abstract
Alzheimer’s disease (AD) is characterized by complex interactions between neuropathological markers, metabolic dysregulation, and structural brain changes. In this study, we utilized a multimodal approach, combining immunohistochemistry, functional metabolic mapping, and microstructure sensitive diffusion MRI (dMRI) to progressively investigate these interactions in the [...] Read more.
Alzheimer’s disease (AD) is characterized by complex interactions between neuropathological markers, metabolic dysregulation, and structural brain changes. In this study, we utilized a multimodal approach, combining immunohistochemistry, functional metabolic mapping, and microstructure sensitive diffusion MRI (dMRI) to progressively investigate these interactions in the 5xFAD mouse model of AD. Our analysis revealed age-dependent and region-specific accumulation of key AD markers, including amyloid-beta (Aβ), GFAP, and IBA1, with significant differences observed between the hippocampal formation and upper and lower regions of the cortex by 6 months of age. Functional metabolic mapping validated localized disruptions in energy metabolism, with glucose hypometabolism in the hippocampus and impaired astrocytic metabolism in the cortex. Notably, increased cortical glutaminolysis suggested a shift in microglial metabolism, reflecting an adaptive response to neuroinflammatory processes. While dMRI showed no significant microstructural differences between 5xFAD and wild-type controls, the study highlights the importance of metabolic alterations as critical events in AD pathology. These findings emphasize the need for targeted therapeutic strategies addressing specific metabolic disturbances and underscore the potential of integrating advanced imaging with metabolic and molecular analyses to advance our understanding of AD progression. Full article
Show Figures

Figure 1

Figure 1
<p>Progressive increase in Aβ accumulation, myelin degeneration, and gliosis in the 5xFAD brains. Representative fluorescence staining images from triple-stainings in brain slices of 5xFAD and WT with (<b>A</b>) Aβ/GFAP/DAPI, (<b>B</b>) IBA1/GFAP/DAPI, and (<b>C</b>) Aβ/MBP/DAPI at 2 (2M) and 6 (6M) month timepoints. (1) 5× magnification (2.02 µm/pixel) microscopy images of brain slices showing subiculum, upper cortex, lower cortex, and white matter. (2) 20× magnification (0.5128 µm/pixel) microscopy images of brain slices showing subiculum. Scale bars indicate 500 µm and 100 µm for upper and lower rows, respectively.</p>
Full article ">Figure 2
<p>Quantification of pathological markers in the brains of 5xFAD and WT mice at two ages. Fluorescence microscopy images (represented in <a href="#biomolecules-14-01294-f001" class="html-fig">Figure 1</a>) were quantified, and the corresponding signal derived from the markers (<b>A</b>) Aβ, (<b>B</b>) GFAP, (<b>C</b>) IBA1 and (<b>D</b>) MBP is presented. Four different brain regions are shown: (1) subiculum, (2) upper cortex, (3) lower cortex, and (4) white matter. Each bar graph shows populations of the level of markers in 2M 5xFAD, 2M WT, 6M 5xFAD, and 6M WT mice. The bar graphs are presented as mean percentages of either positive pixels (<b>A</b>–<b>C</b>), or mean gray-scale value (<b>D</b>) ± SD. The number of brain slices is 6 (from 3 independent mice per genotype per age). The statistical comparisons (determined with ANOVA and Tukey’s test) are presented with <span class="html-italic">p</span> values above the bars, with the significant values in bold.</p>
Full article ">Figure 3
<p>Cortical acetate metabolism and hippocampal glucose metabolism is selectively affected in 6M 5xFAD brains. (<b>A</b>) Mitochondrial oxidative metabolism of [U-<sup>13</sup>C]glucose (glc, left axes orange bars) or [1,2-<sup>13</sup>C]acetate (ace, right axes, green bars) gives rise to <sup>13</sup>C-enrichment (detected by GC–MS) in TCA cycle intermediates in acutely isolated slices from (<b>B</b>) cerebral cortex or (<b>C</b>) hippocampus incubated with the labeled substrates for 60 min. Molecular carbon labeling (MCL), the weighted average of the carbon labeling in given metabolic intermediates, is presented. [U-<sup>13</sup>C]glucose reflects overall energy metabolism where neurons are the main energy consumers, while [1,2-<sup>13</sup>C]acetate is predominantly metabolized in astrocytes. In the cerebral cortex, overall maintained <sup>13</sup>C-incorporation in TCA cycle metabolites from [U-<sup>13</sup>C]glucose was observed, while lower MCL in intermediates from [1,2-<sup>13</sup>C]acetate was found in 5xFAD mice compared to wild-type (WT) controls. The opposite was observed for the hippocampal slices, where a lower MCL in intermediates resulting from [U-<sup>13</sup>C]glucose metabolism but maintained [1,2-<sup>13</sup>C]acetate was detected in the 5xFAD mice vs. WT. Values represent mean (±) SD (n  =  4 animals). WT animals are represented as circles, while 5xFAD animals are represented as triangles. The statistical significance (determined with Student’s unpaired <span class="html-italic">t</span>-test) is presented with <span class="html-italic">p</span> values above the bars, with significant values in bold.</p>
Full article ">Figure 4
<p>Amino acids derived from acetate metabolism are lower in the cortex but unchanged in the hippocampus in 6M 5xFAD brains. (<b>A</b>) Mitochondrial oxidative metabolism of [U-<sup>13</sup>C]glucose (glc, left axes, orange bars) or [1,2-<sup>13</sup>C]acetate (ace, right axes, green bars) gives rise to <sup>13</sup>C-enrichment (detected by GC–MS) in TCA cycle intermediates and derived amino acids in acutely isolated slices from (<b>B</b>) cerebral cortex or (<b>C</b>) hippocampus incubated with the labeled substrates for 60 min. Molecular carbon labeling (MCL), the weighted average of the carbon labeling in given metabolic intermediates, is presented. [U-<sup>13</sup>C]glucose reflects overall energy metabolism where neurons are the main energy consumers, while [1,2-<sup>13</sup>C]acetate is predominantly metabolized in astrocytes. In the cerebral cortex, overall maintained <sup>13</sup>C-incorporation in TCA cycle-derived amino acids from [U-<sup>13</sup>C]glucose was observed, while lower MCL in amino acids from [1,2-<sup>13</sup>C]acetate metabolism was found in 5xFAD mice compared to wild-type (WT) controls. In hippocampal slices, a maintained MCL in amino acids resulting from [U-<sup>13</sup>C]glucose and [1,2-<sup>13</sup>C]acetate metabolism was detected in the 5xFAD mice vs. WT. Values represent mean (±) SD (n  =  4 animals). WT animals are represented as circles, while 5xFAD animals are represented as triangles. The statistical significance (determined with Student’s unpaired <span class="html-italic">t</span>-test) is presented with <span class="html-italic">p</span> values above the bars, with significant values in bold.</p>
Full article ">Figure 5
<p>Glutamine uptake is unchanged while its metabolism is higher in the 5xFAD cortex. (<b>A</b>) Uptake and metabolism of [U-<sup>13</sup>C]glutamine (gln) gives rise to <sup>13</sup>C-enrichment (detected by GC–MS) in glutamate, GABA, and TCA cycle intermediates in acutely isolated slices from (<b>B</b>) cerebral cortex or (<b>C</b>) hippocampus incubated with the labeled substrates for 60 min. <sup>13</sup>C-enrichment from direct glutamine metabolism is presented as M + X labeling % (X = number of <sup>13</sup>C-carbons in a given molecule). In the cerebral cortex, overall maintained <sup>13</sup>C-incorporation in amino acids from [U-<sup>13</sup>C]gln was observed, while higher labeling was detected in most TCA cycle intermediates in 5xFAD mice compared to wild-type (WT) controls, suggesting increased glutaminolysis. In hippocampal slices, a maintained labeling in amino acids and TCA cycle intermediates resulting from [U-<sup>13</sup>C]gln metabolism, with the exception of fumarate, was detected in the 5xFAD mice vs. WT. Values represent mean (±) SD (n  =  4 animals). WT animals are represented as circles, while 5xFAD animals are represented as triangles. The statistical significance (determined with Student’s unpaired <span class="html-italic">t</span>-test) is presented with <span class="html-italic">p</span> values above the bars, with significant values in bold.</p>
Full article ">Figure 6
<p>The average mean diffusivity (MD) within each region of interest (ROI) was decreased in 5xFAD brains of both age groups. The MD was found consistently decreased in all ROIs of 5xFAD brains compared to WT brains. However, this difference between genotypes was not statistically significant. Additionally, while a small age-related increase in MD was observed in all ROIs for both genotypes, this difference was mainly evident in 5xFAD brains. Group means with 95% confidence intervals are superimposed on the individual observations. 2M = 2-month-old, 6M = 6-month-old. Sub = subiculum, CA = cornu ammonis of the hippocampal formation, hip = hippocampus, CC = corpus callosum, WM = white matter, LC = lower cortex, UC = upper cortex, and neo = neocortex.</p>
Full article ">Figure 7
<p>6M 5xFAD brains exhibited decreased averaged mean kurtosis (MK) in all regions of interest (ROIs). While the differences in MK between genotypes of 2-month-old (2M) brains varied between ROIs, a small but consistent decrease was observed in the 6-month-old (6M) 5xFAD brains compared to WT brains. This difference was not statistically significant. An age-related increase was observed in the ROIs of both genotypes, although less pronounced in 5xFAD brains. This age difference was not statistically different either. Group means with 95% confidence intervals are superimposed on the individual observations. Sub = subiculum, CA = cornu ammonis of the hippocampal formation, hip = hippocampus, CC = corpus callosum, WM = white matter, LC = lower cortex, UC = upper cortex, and neo = neocortex.</p>
Full article ">Figure 8
<p>The relative volume of all ROIs was unchanged in 5xFAD brains. No difference was observed in the relative volume of the ROIs between genotypes. A statistically significant age × ROI interaction was observed. However, the simple main effects analysis found no statistically significant effect of age in any of the ROIs. The dotted line indicates that LC, UC, and neo are plotted against the right y-axis. Group means with 95% confidence intervals are superimposed on the individual observations. 2M = 2-month-old, 6M = 6-month-old. Sub = subiculum, CA = cornu ammonis of the hippocampal formation, hip = hippocampus, CC = corpus callosum, WM = white matter, LC = lower cortex, UC = upper cortex, and neo = neocortex.</p>
Full article ">
Back to TopTop