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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
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

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

Search Results (1,989)

Search Parameters:
Keywords = degenerative diseases

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 4678 KiB  
Article
Deep Learning-Based Diagnosis Algorithm for Alzheimer’s Disease
by Zhenhao Jin, Junjie Gong, Minghui Deng, Piaoyi Zheng and Guiping Li
J. Imaging 2024, 10(12), 333; https://doi.org/10.3390/jimaging10120333 - 23 Dec 2024
Abstract
Alzheimer’s disease (AD), a degenerative condition affecting the central nervous system, has witnessed a notable rise in prevalence along with the increasing aging population. In recent years, the integration of cutting-edge medical imaging technologies with forefront theories in artificial intelligence has dramatically enhanced [...] Read more.
Alzheimer’s disease (AD), a degenerative condition affecting the central nervous system, has witnessed a notable rise in prevalence along with the increasing aging population. In recent years, the integration of cutting-edge medical imaging technologies with forefront theories in artificial intelligence has dramatically enhanced the efficiency of identifying and diagnosing brain diseases such as AD. This paper presents an innovative two-stage automatic auxiliary diagnosis algorithm for AD, based on an improved 3D DenseNet segmentation model and an improved MobileNetV3 classification model applied to brain MR images. In the segmentation network, the backbone network was simplified, the activation function and loss function were replaced, and the 3D GAM attention mechanism was introduced. In the classification network, firstly, the CA attention mechanism was added to enhance the model’s ability to capture positional information of disease features; secondly, dilated convolutions were introduced to extract richer features from the input feature maps; and finally, the fully connected layer of MobileNetV3 was modified and the idea of transfer learning was adopted to improve the model’s feature extraction capability. The results of the study showed that the proposed approach achieved classification accuracies of 97.85% for AD/NC, 95.31% for MCI/NC, 93.96% for AD/MCI, and 92.63% for AD/MCI/NC, respectively, which were 3.1, 2.8, 2.6, and 2.8 percentage points higher than before the improvement. Comparative and ablation experiments have validated the proposed classification performance of this method, demonstrating its capability to facilitate an accurate and efficient automated auxiliary diagnosis of AD, offering a deep learning-based solution for it. Full article
(This article belongs to the Special Issue Advancements in Artificial Intelligence for Medical Images)
Show Figures

Figure 1

Figure 1
<p>Brain MR image preprocessing.</p>
Full article ">Figure 2
<p>DAGAN structure.</p>
Full article ">Figure 3
<p>The workflow flowchart of 3D GAM.</p>
Full article ">Figure 4
<p>Improved 3D DenseNet model structure.</p>
Full article ">Figure 5
<p>Improved MobileNetV3 structure.</p>
Full article ">Figure 6
<p>AD automatic auxiliary diagnosis algorithm based on improved MobileNetV3.</p>
Full article ">Figure 7
<p>Local magnification and comparison of segmentation slices.</p>
Full article ">Figure 8
<p>Comparison of segmentation results on ADNI dataset with different methods.</p>
Full article ">Figure 9
<p>AD/NC confusion matrices before and after model improvement.</p>
Full article ">Figure 10
<p>AD/NC ROC curve before and after model improvement.</p>
Full article ">Figure 11
<p>MCI/NC confusion matrices before and after model improvement.</p>
Full article ">Figure 12
<p>MCI/NC ROC curve before and after model improvement.</p>
Full article ">Figure 13
<p>AD/MCI confusion matrices before and after model improvement.</p>
Full article ">Figure 14
<p>AD/MCI ROC curve before and after model improvement.</p>
Full article ">Figure 15
<p>AD/MCI/NC confusion matrices before and after model improvement.</p>
Full article ">Figure 16
<p>AD/MCI/NC ROC curve before and after model improvement.</p>
Full article ">
22 pages, 1090 KiB  
Review
Modulating Autophagy in Osteoarthritis: Exploring Emerging Therapeutic Drug Targets
by Corina Andrei, Dragos Paul Mihai, George Mihai Nitulescu, Georgiana Nitulescu and Anca Zanfirescu
Int. J. Mol. Sci. 2024, 25(24), 13695; https://doi.org/10.3390/ijms252413695 - 21 Dec 2024
Viewed by 355
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by the breakdown of cartilage and the subsequent inflammation of joint tissues, leading to pain and reduced mobility. Despite advancements in symptomatic treatments, disease-modifying therapies for OA remain limited. This narrative review examines the dual [...] Read more.
Osteoarthritis (OA) is a degenerative joint disease characterized by the breakdown of cartilage and the subsequent inflammation of joint tissues, leading to pain and reduced mobility. Despite advancements in symptomatic treatments, disease-modifying therapies for OA remain limited. This narrative review examines the dual role of autophagy in OA, emphasizing its protective functions during the early stages and its potential to contribute to cartilage degeneration in later stages. By delving into the molecular pathways that regulate autophagy, this review highlights its intricate interplay with oxidative stress and inflammation, key drivers of OA progression. Emerging therapeutic strategies aimed at modulating autophagy are explored, including pharmacological agents such as AMP kinase activators, and microRNA-based therapies. Preclinical studies reveal encouraging results, demonstrating that enhancing autophagy can reduce inflammation and decelerate cartilage degradation. However, the therapeutic benefits of autophagy modulation depend on precise, stage-specific approaches. Excessive or dysregulated autophagy in advanced OA may lead to chondrocyte apoptosis, exacerbating joint damage. This review underscores the promise of autophagy-based interventions in bridging the gap between experimental research and clinical application. By advancing our understanding of autophagy’s role in OA, these findings pave the way for innovative and effective therapies. Nonetheless, further research is essential to optimize these strategies, address potential off-target effects, and develop safe, targeted treatments that improve outcomes for OA patients. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Key signaling pathways associated with autophagy regulation in OA. AMPK, AMP-activated protein kinase; AKT, Protein kinase B; Beclin1, Autophagy-related protein; IL-1β, Interleukin 1 beta; IL-15, Interleukin 15; IL-6, Interleukin 6; LC3, Microtubule-associated proteins 1A/1B light chain 3; LC3I/LC3II, Forms of LC3 during autophagy; miR-100-5p, MicroRNA-100-5p; miR-155, MicroRNA-155; mTORC1, Mechanistic target of rapamycin complex 1; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; P53, Tumor protein P53; PI3K, Phosphoinositide 3-kinases; PP2AC, Protein phosphatase 2A catalytic subunit; RAG GTPase, Ras-related GTPase; REDD1, regulated in development and DNA damage responses 1; ROS, reactive oxygen species; TNFα, Tumor necrosis factor alpha; TSC1/2, Tuberous sclerosis complex 1/2; ULK1, Unc-51-like autophagy activating kinase 1; →, stimulation; <span class="html-fig-inline" id="ijms-25-13695-i001"><img alt="Ijms 25 13695 i001" src="/ijms/ijms-25-13695/article_deploy/html/images/ijms-25-13695-i001.png"/></span>, inhibition.</p>
Full article ">
15 pages, 618 KiB  
Article
Is Ultrasonography an Effective Method for Diagnosing Degenerative Changes in the Temporomandibular Joint?
by Barbara Wojciechowska, Arkadiusz Szarmach, Adam Michcik, Maciej Sikora and Barbara Drogoszewska
Biomedicines 2024, 12(12), 2915; https://doi.org/10.3390/biomedicines12122915 - 21 Dec 2024
Viewed by 221
Abstract
Background: The accurate diagnosis of degenerative joint diseases (DJDs) of the temporomandibular joint (TMJ) presents a significant clinical challenge due to their progressive nature and the complexity of associated structural changes. These conditions, characterized by cartilage degradation, subchondral bone remodeling, and eventual joint [...] Read more.
Background: The accurate diagnosis of degenerative joint diseases (DJDs) of the temporomandibular joint (TMJ) presents a significant clinical challenge due to their progressive nature and the complexity of associated structural changes. These conditions, characterized by cartilage degradation, subchondral bone remodeling, and eventual joint dysfunction, necessitate reliable and efficient imaging techniques for early detection and effective management. Cone-beam computed tomography (CBCT) is widely regarded as the gold standard for evaluating osseous changes in the TMJ, offering detailed visualization of bony structures. However, ultrasonography (US) has emerged as a promising alternative, offering a non-invasive and radiation-free option for assessing TMJ disorders. This study aims to evaluate the diagnostic accuracy of US in identifying degenerative changes in the TMJ, with CBCT serving as the definitive diagnostic reference. By analyzing the sensitivity, specificity, and predictive values of US in detecting key degenerative markers—such as subchondral erosion, osteophytes, and joint space narrowing—this investigation seeks to assess its utility as a screening tool and its potential integration into clinical workflows. Methods: Forty adult patients presenting temporomandibular joint disorders were included in our cross-sectional study. Each patient underwent a clinical examination and was subjected to cone-beam computed tomography (CBCT) and ultrasonography (US). A statistical analysis was performed to compare the imaging results from CBCT and US. Results: The results are summarized in three tables. The first table presents a comparative analysis of radiological outcomes in patients with temporomandibular joint disorders using different imaging techniques. CBCT demonstrated higher sensitivity in detecting osteophytes in the right mandibular head (27.50% vs. 7.50%, p = 0.027) and higher detection rates for erosions, though without a significant advantage over US. The second table analyzes the consistency of diagnostic results between CBCT and US. A moderate agreement was observed for detecting normal bone structures, with AC1 values of 0.58 for the right and 0.68 for the left mandibular head (p < 0.001). The third table evaluates the diagnostic accuracy of US compared to CBCT. US demonstrated a positive predictive value (PPV) of 90% for detecting normal conditions, indicating its high reliability as a screening tool for normal findings. US demonstrates higher effectiveness in ruling out certain issues due to its high specificity and negative predictive value. However, its lower sensitivity in detecting abnormalities may lead to both false-positive and false-negative results. Conclusions: US holds significant promise as a screening modality for detecting normal anatomical features of the temporomandibular joint, its limitations in identifying more complex degenerative changes necessitate a cautious and integrated approach to TMJ diagnostics. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) CBCT (coronal view). Black arrow points to erosion and subchondral cyst. Anatomical structures of the TMJ: 1. Disc, 2. Condyle, 3. Temporal Bone; (<b>b</b>) CBCT (sagittal view). Black arrow indicates osteophyte. Anatomical structures of the TMJ: 1. Disc, 2. Condyle, 3. Temporal Bone.</p>
Full article ">Figure 2
<p>(<b>a</b>) Ultrasonography (sagittal view). Black arrow is pointing to erosion. Anatomical structures: 1. Glenoid Fossa, 2. Disc, 3. Condyle, 4. Masseter muscle; (<b>b</b>) Ultrasonography (axial view). Black arrow shows osteophyte. Anatomical structures: 1. Glenoid Fossa, 2. Disc, 3. Condyle, 4. Masseter muscle.</p>
Full article ">
14 pages, 1931 KiB  
Article
Micro-CT Assessment During Embedding of Prototype Ti Alloy Multi-Spiked Connecting Scaffold in Subchondral Trabecular Bone of Osteoarthritic Femoral Heads, Depending on Host BMI
by Ryszard Uklejewski, Mariusz Winiecki, Adam Patalas, Patryk Mietliński, Paweł Zawadzki and Mikołaj Dąbrowski
J. Funct. Biomater. 2024, 15(12), 387; https://doi.org/10.3390/jfb15120387 - 21 Dec 2024
Viewed by 439
Abstract
The prototype of a biomimetic multi-spiked connecting scaffold (MSC-Scaffold) represents an essential innovation in the fixation in subchondral trabecular bone of components for a new generation of entirely cementless hip resurfacing arthroplasty (RA) endoprostheses. In designing such a functional biomaterial scaffold, identifying the [...] Read more.
The prototype of a biomimetic multi-spiked connecting scaffold (MSC-Scaffold) represents an essential innovation in the fixation in subchondral trabecular bone of components for a new generation of entirely cementless hip resurfacing arthroplasty (RA) endoprostheses. In designing such a functional biomaterial scaffold, identifying the microstructural and mechanical properties of the host bone compromised by degenerative disease is crucial for proper post-operative functioning and long-term maintenance of the endoprosthesis components. This study aimed to explore, depending on the occurrence of obesity, changes in the microstructure and mechanical properties of the subchondral trabecular bone in femoral heads of osteoarthritis (OA) patients caused by the MSC-Scaffold embedding. Computed microtomography (micro-CT) scanning of femoral heads from OA patients was conducted before and after the mechanical embedding of the MSC-Scaffold. Bone morphometric parameters such as bone volume/total volume (BV/TV), trabecular thickness (Tb.Th), and trabecular number (Tb.N) for regions surrounding the MSC-Scaffold were computed, and the mechanical properties such as bone density (ρB), bone compressive strength (S), and the Young’s modulus (E) within these regions were calculated. A statistically significant increase in BV/TV (by 15.0% and 24.9%) and Tb.Th (by 13.1% and 42.5%) and a decrease in Tb.N (by 15.2% and 23.6%) were observed, which translates to an increase in ρB (by 15.0% and 24.9%), S (by 28.8% and 49.5%), and E (by 18.0% and 29.8%) in non-obese patients and obese patients, respectively. These changes in properties are favorable for the mechanical loads’ transfer from the artificial joint surface via the MSC-Scaffold to the periarticular trabecular bone of the OA femoral head in the postoperative period. Full article
(This article belongs to the Special Issue Functional Scaffolds for Bone and Joint Surgery)
Show Figures

Figure 1

Figure 1
<p>The representative (<b>a</b>) micro-CT 3D reconstruction and (<b>b</b>) cross-section of the OA femoral head specimen with the initially embedded MSC-Scaffold (cylindrical-shaped ROI marked with a green line); (<b>c</b>) the magnified fragment of the cross-section revealing the densified bone region in the peri-implant subchondral trabecular bone within which the spatial extent of subchondral trabecular bone densification due to the MSC-Scaffold prototype initial embedding (red line) and the cuboid-shaped ROI being the subject of morphometric measurements (orange line) are marked.</p>
Full article ">Figure 2
<p>Averaged changes in subchondral trabecular bone relative area (BA/TA) values at different levels below the reference plane due to the initial embedding of the MSC-Scaffold in femoral head specimens from (<b>a</b>) NOP and (<b>b</b>) OP with OA.</p>
Full article ">Figure 3
<p>Bone morphometric parameters of subchondral trabecular bone in cuboid-shaped ROI femoral heads of OA patients before and after mechanical embedding of the MSC-Scaffold: (<b>a</b>) Bone volume/total volume (BV/TV); (<b>b</b>) Trabecular thickness (Tb.Th), and (<b>c</b>) Trabecular number (Tb.N). Data is presented as a mean ± standard deviation. Probability values (<span class="html-italic">p</span>-values) from the performed <span class="html-italic">t</span>-test between the results are given above the bars.</p>
Full article ">Figure 4
<p>The values of the mechanical properties of subchondral trabecular bone before and after initial embedding of the MSC-Scaffold in femoral head specimens from NOP and OP with OA including (<b>a</b>) subchondral trabecular bone density <span class="html-italic">ρ<sub>B</sub></span>, (<b>b</b>) compressive strength <span class="html-italic">S</span>, and (<b>c</b>) Young’s modulus <span class="html-italic">E</span> calculated from Formulas (1)–(3). Data are presented as a mean ± standard deviation. Probability values (<span class="html-italic">p</span>-values) from the performed <span class="html-italic">t</span>-test between the results are given above the bars.</p>
Full article ">
16 pages, 11207 KiB  
Article
IGF-1 Signaling Modulates Oxidative Metabolism and Stress Resistance in ARPE-19 Cells Through PKM2 Function
by Silvia Ravera, Alessandra Puddu, Nadia Bertola, Daniela Verzola, Elisa Russo, Davide Maggi and Isabella Panfoli
Int. J. Mol. Sci. 2024, 25(24), 13640; https://doi.org/10.3390/ijms252413640 - 20 Dec 2024
Viewed by 158
Abstract
The retinal pigment epithelium (RPE) contributes to retinal homeostasis, and its metabolic dysfunction is implied in the development of retinal degenerative disease. The isoform M2 of pyruvate kinase (PKM2) is a key factor in cell metabolism, and its function may be affected by [...] Read more.
The retinal pigment epithelium (RPE) contributes to retinal homeostasis, and its metabolic dysfunction is implied in the development of retinal degenerative disease. The isoform M2 of pyruvate kinase (PKM2) is a key factor in cell metabolism, and its function may be affected by insulin-like growth factor 1 (IGF-1). This study aims to investigate the effect of IGF-1 on PKM2 modulation of RPE cells and whether co-treatment with klotho may preserve it. ARPE-19 cells, an ex vivo model of human pigmented epithelium, were exposed to IGF-1. Then, we evaluated PKM2 expression, dimerization and subcellular localization, energy metabolism, and redox balance, and whether pre-treatment with Klotho may antagonize the effects of IGF-1. The results show that IGF-1 favors PKM2 dimerization, thus reducing the activity of PKM2 and leading to an altered cellular energy status coupled with reduced oxidative stress. In conclusion, PKM2 plays a pivotal role in the modulation of RPE metabolism and redox balance and could explain the mechanisms through which IGF-1 participates in the pathogenesis of some retinal diseases. Klotho may exert protective effects by mitigating the IGF-1 signal and its effect on mitochondrial function. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>IGF-1 increases the expression and dimerization of the isoform M2 of pyruvate kinase (PKM2) in ARPE-19. (<b>A</b>) Protein expression levels of PKM2 in ARPE-19 cells treated with IGF-1 for 2, 4, and 24 h. (<b>B</b>) Analysis of the different structural forms of PKM2 in ARPE-19 cells treated with IGF-1 for 2, 4, and 24 h. Data are representative of three independent experiments (n = 3) and are indicated as means ± SD. Statistical analysis was performed with one-way ANOVA followed by Tukey’s multiple comparisons test. *, **, and *** indicate <span class="html-italic">p</span> &lt; 0.05, 0.01, or 0.0001, respectively.</p>
Full article ">Figure 2
<p>Representative immunofluorescence images of the M2 isoform of pyruvate kinase (PKM2) in ARPE-19 cells treated with IGF-1 for 4 and 24 h (n = 3). The red signal represents the immunolocalization of PKM2 during IGF-1 treatment, while cell nuclei are stained blue with DAPI. White scale bars correspond to 15 μm. The higher-magnification images correspond to the areas enclosed by white squares for each condition in the merged-signals column. The white arrows indicate the nuclear localization of PKM2.</p>
Full article ">Figure 3
<p>Pyruvate kinase (PK) activity in ARPE-19 cells treated with IGF-1 in the absence or presence of Klotho. The graph shows the PK activity in ARPE-19 cells treated with IGF-1 for 2, 4, and 24 h in the absence or presence of Klotho. Data are representative of four independent experiments (n = 4) and are indicated as means ± SD. Statistical analysis was performed with one-way ANOVA followed by Tukey’s multiple comparisons test. * and **** indicate <span class="html-italic">p</span> &lt; 0.05 or 0.0001, respectively.</p>
Full article ">Figure 4
<p>Evaluation of ATP synthesis and oxygen consumption rate (OCR) as markers of OxPhos activity in ARPE-19 cells treated with IGF-1 in the absence or presence of Klotho. All data were obtained from ARPE-19 cells treated with IGF-1 for 2, 4, and 24 h in the absence or presence of Klotho. (<b>A</b>) ATP synthesis through F<sub>o</sub>F<sub>1</sub> ATP synthase; (<b>B</b>) OCR; percentages of ATP synthesis (<b>C</b>) and OCR (<b>D</b>) sensitive to BPTES (a glutaminase inhibitor used to assess metabolic dependence on glutamine), Etomoxir (a CTP1-A inhibitor used to assess metabolic dependence on fatty acid), and UK5099 (a mitochondrial pyruvate carrier inhibitor used to assess metabolic dependence on pyruvate and consequently on glucose) to evaluate the dependency on energy substrates. For each panel, data are representative of four independent experiments (n = 4) and are indicated as means ± SD. Statistical analysis was performed with one-way ANOVA followed by Tukey’s multiple comparisons test. In panels A and B, *** and **** indicate <span class="html-italic">p</span> &lt; 0.001 or 0.0001, respectively. In panels C and D, **, ***, and **** indicate <span class="html-italic">p</span> &lt; 0.01, 0.001, or 0.0001, respectively, between untreated cells and cells treated with IGF-1 for 24 h.</p>
Full article ">Figure 5
<p>Anaerobic glycolytic metabolism in ARPE-19 cells treated with IGF-1 in the absence or presence of Klotho. All data were obtained from ARPE-19 cells treated with IGF-1 for 2, 4, and 24 h in the absence or presence of Klotho. (<b>A</b>) Lactate dehydrogenase (LDH) activity. (<b>B</b>) Glycolytic yield. Data are representative of four independent experiments (n = 4) and are indicated as means ± SD. For Panel A, statistical analysis was performed with one-way ANOVA followed by Tukey’s multiple comparisons test; for Panel B, statistical analysis was performed by a <span class="html-italic">t</span>-test for each time point. ** and **** indicate <span class="html-italic">p</span> &lt; 0.01 or 0.0001, respectively.</p>
Full article ">Figure 6
<p>Cellular energy status of ARPE-19 cells treated with IGF-1 in the absence or presence of Klotho. All data were obtained from ARPE-19 cells treated with IGF-1 for 2, 4, and 24 h in the absence or presence of Klotho. (<b>A</b>) ATP intracellular concentration. (<b>B</b>) AMP intracellular concentration. (<b>C</b>) ATP/AMP ratio as a cellular energy status marker. Data are representative of four independent experiments (n = 4) and are indicated as means ± SD. Statistical analysis was performed with one-way ANOVA followed by Tukey’s multiple comparisons test. *, **, ***, and **** indicate <span class="html-italic">p</span> &lt; 0.05, 0.01, 0.001, or 0.0001, respectively.</p>
Full article ">Figure 7
<p>Lipid peroxidation accumulation and glucose-6-phosphate dehydrogenase (G6PD), glutathione reductase (GR), and glutathione peroxidase (GPx) activity in ARPE-19 cells treated with IGF-1 in the absence or presence of Klotho. All data were obtained from ARPE-19 cells treated with IGF-1 for 2, 4, and 24 h in the absence or presence of Klotho. (<b>A</b>) Malondialdehyde (MDA) intracellular concentration as a lipid peroxidation marker. (<b>B</b>) G6PD activity. (<b>C</b>) GR activity. (<b>D</b>) GPx activity. Data are representative of four independent experiments (n = 4) and are indicated as means ± SD. Statistical analysis was performed with one-way ANOVA followed by Tukey’s multiple comparisons test. *, **, ***, and **** indicate <span class="html-italic">p</span> &lt; 0.05, 0.01, 0.001, or 0.0001, respectively.</p>
Full article ">Figure 8
<p>Intracellular signaling of IGF-1 in ARPE-19 cells. All data were obtained from ARPE-19 cells serum-starved overnight, then exposed to 100 nmol/L IGF-1 for 5 or 20 min in the absence or presence of Klotho. Then, cells were lysed and immunoblotted with anti-phospho antibodies against IGF1R (<b>A</b>), IRS-1 (<b>B</b>), and AKT (<b>C</b>). Representative Western blotting and the quantification of densitometries of Western blot bands are shown. Data are expressed as mean ± SD of fold induction relative to GAPDH (n = 3). Statistical analysis was performed with one-way ANOVA followed by Tukey’s multiple comparisons test. *, **, ***, and **** indicate <span class="html-italic">p</span> &lt; 0.05, 0.01, 0.001, or 0.0001, respectively.</p>
Full article ">Figure 9
<p>Subcellular localization of isoform M2 of pyruvate kinase (PKM2) in ARPE-19 cells treated with IGF-1 in the absence or presence of Klotho. ARPE-19 cells were treated with IGF-1 for 2, 4, and 24 h in the absence or presence of Klotho. Then, subcellular fractions of cells were obtained ((<b>A</b>) cytosol; (<b>B</b>) nuclear soluble; (<b>C</b>) chromatin bound). Representative Western blotting and quantification of the densitometries of Western blot bands are shown. Data are expressed as mean ± SD of fold induction relative to the amounts of each respective loading control (n = 3). Statistical analysis was performed with one-way ANOVA followed by Tukey’s multiple comparisons test. *, **, ***, and **** indicate <span class="html-italic">p</span> &lt; 0.05, 0.01, 0.001, or 0.0001, respectively.</p>
Full article ">
22 pages, 12235 KiB  
Article
The Focal Induction of Reactive Oxygen Species in Rats as a Trigger of Aortic Valve Degeneration
by Jessica Isabel Selig, Yukiharu Sugimura, Shintaro Katahira, Marco Polidori, Laura Alida Jacobi, Olga Medovoj, Sarah Betke, Mareike Barth, Artur Lichtenberg, Payam Akhyari and Jan-Philipp Minol
Antioxidants 2024, 13(12), 1570; https://doi.org/10.3390/antiox13121570 - 20 Dec 2024
Viewed by 273
Abstract
Background: Degenerative aortic valve disease (DAVD) is a multifactorial process. We developed an animal model to analyze the isolated, local effect of reactive oxygen species (ROS) on its pathophysiology. Methods: We utilized a photodynamic reaction (PDR) as a source of ROS in the [...] Read more.
Background: Degenerative aortic valve disease (DAVD) is a multifactorial process. We developed an animal model to analyze the isolated, local effect of reactive oxygen species (ROS) on its pathophysiology. Methods: We utilized a photodynamic reaction (PDR) as a source of ROS in the aortic valve by aiming a laser at the aortic valve for 60 min after the administration of a photosensitizer 24 h prior. ROS, laser, and sham groups (n = 7 each) for every observation period (t = 0; t = 8 d; t = 84 d; t = 168 d) were established. The amount of ROS generation; morphological changes; inflammatory, immune, and apoptotic reactions; and hemodynamic changes in the aortic valves were assessed using appropriate histological, immunohistological, immunohistochemical, and echocardiographic methods. Results: The ROS group displayed an increased amount of ROS (p < 0.01) and increased inflammatory activation of the endothelium (p < 0.05) at t = 0. In the ROS group, aortic valves were calcified (p < 0.05) and the transvalvular gradient was increased (p < 0.01) at t = 168 d. Conclusion: The small animal model employed here may serve as a platform for analyzing ROS’s isolated role in the DAVD context. Full article
(This article belongs to the Special Issue Redox Regulation in Cardiovascular Diseases)
Show Figures

Figure 1

Figure 1
<p>For the ROS, laser, and sham groups, a laser fiber was inserted under echocardiographic control. (<b>a</b>) The label “1” in (<b>b</b>) shows the fiber; the circle in (<b>b</b>) indicates the aortic valve. The label “2” in (<b>b</b>) indicates the left ventricle. (<b>c</b>) A typical specimen of the aortic valve in the aortic root with the adjacent parts of the myocardium and the aorta.</p>
Full article ">Figure 2
<p>Indirect ROS detection by 8-OHdG fluorescence. After the initial procedure, the content of generated ROS was assessed indirectly via 8-OHdG fluorescence at t = 0 and t = 8 d ((<b>a</b>,<b>d</b>) = ROS; (<b>b</b>,<b>e</b>) = laser; (<b>c</b>,<b>f</b>) = sham at t = 0). Quantitative analysis of 8-OHdG (mean gray value normalized to the positive control of the staining run) displayed an increase in the laser and ROS groups at t = 0, with a significant difference between the ROS and the sham group (<b>g</b>). On the contrary, no differences between the groups were found at t = 8 d (<b>h</b>). 8-OHdG (red) with DAPI counterstain (blue); scale bars = 50 µm. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Analyses of the cleaved-caspase-3 activity. Neither the cross-section images of the ROS groups at t = 0 (<b>a</b>) and t = 8 d (<b>b</b>) nor those of the corresponding laser (<b>c</b>,<b>d</b>) and sham groups (<b>e</b>,<b>f</b>) displayed distinctive cleaved-caspase-3 activity. Immunohistochemistry: cleaved-caspase-3 (DAB, brown) with hemalum counterstain (blue); scale bars = 1000 µm.</p>
Full article ">Figure 4
<p>Concentration of monocytes was quantitatively assessed via CD 68 expression. For this purpose, the percentage of CD 68-specific signals in the total tissue area was calculated. Representative cross-section images at t = 8 d (ROS group: (<b>a</b>,<b>c</b>) = annulus, (<b>b</b>,<b>d</b>) = cusp; laser group: (<b>e</b>,<b>g</b>) = annulus; (<b>f</b>,<b>h</b>) = cusp; sham group: (<b>i</b>,<b>k</b>) = annulus; (<b>j</b>,<b>l</b>) = cusp). Quantitative analysis (<b>m</b>–<b>p</b>). CD 68 (red) with DAPI counterstain (blue); scale bars = 50 µm.</p>
Full article ">Figure 4 Cont.
<p>Concentration of monocytes was quantitatively assessed via CD 68 expression. For this purpose, the percentage of CD 68-specific signals in the total tissue area was calculated. Representative cross-section images at t = 8 d (ROS group: (<b>a</b>,<b>c</b>) = annulus, (<b>b</b>,<b>d</b>) = cusp; laser group: (<b>e</b>,<b>g</b>) = annulus; (<b>f</b>,<b>h</b>) = cusp; sham group: (<b>i</b>,<b>k</b>) = annulus; (<b>j</b>,<b>l</b>) = cusp). Quantitative analysis (<b>m</b>–<b>p</b>). CD 68 (red) with DAPI counterstain (blue); scale bars = 50 µm.</p>
Full article ">Figure 4 Cont.
<p>Concentration of monocytes was quantitatively assessed via CD 68 expression. For this purpose, the percentage of CD 68-specific signals in the total tissue area was calculated. Representative cross-section images at t = 8 d (ROS group: (<b>a</b>,<b>c</b>) = annulus, (<b>b</b>,<b>d</b>) = cusp; laser group: (<b>e</b>,<b>g</b>) = annulus; (<b>f</b>,<b>h</b>) = cusp; sham group: (<b>i</b>,<b>k</b>) = annulus; (<b>j</b>,<b>l</b>) = cusp). Quantitative analysis (<b>m</b>–<b>p</b>). CD 68 (red) with DAPI counterstain (blue); scale bars = 50 µm.</p>
Full article ">Figure 5
<p>Analysis of the VCAM-1 expression. Representative cross-section images of the ROS groups at t = 0 (<b>a</b>,<b>c</b>) and at t = 8 d (<b>b</b>,<b>d</b>) displayed the increased expression of VCAM-1 compared to their respective control groups (laser group at t = 0 (<b>e</b>,<b>g</b>) and t = 8 d (<b>f</b>,<b>h</b>); sham group at t = 0 (<b>i</b>,<b>k</b>) and t = 8 d (<b>j</b>,<b>l</b>)). Semiquantitative analysis of VCAM-1 expression ((<b>m</b>–<b>p</b>); 0 = no expression; 5 = maximum). Immunohistological staining of VCAM-1 (DAB, brown) with hemalum counterstain (blue); scale bars = 1000 µm. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5 Cont.
<p>Analysis of the VCAM-1 expression. Representative cross-section images of the ROS groups at t = 0 (<b>a</b>,<b>c</b>) and at t = 8 d (<b>b</b>,<b>d</b>) displayed the increased expression of VCAM-1 compared to their respective control groups (laser group at t = 0 (<b>e</b>,<b>g</b>) and t = 8 d (<b>f</b>,<b>h</b>); sham group at t = 0 (<b>i</b>,<b>k</b>) and t = 8 d (<b>j</b>,<b>l</b>)). Semiquantitative analysis of VCAM-1 expression ((<b>m</b>–<b>p</b>); 0 = no expression; 5 = maximum). Immunohistological staining of VCAM-1 (DAB, brown) with hemalum counterstain (blue); scale bars = 1000 µm. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Analysis of the endothelial layer. While the cusps (<b>a</b>–<b>l</b>) displayed hardly any remaining endothelial layer (red = vWF) at any timepoint, neither in the ROS (<b>a</b>–<b>d</b>) nor in the laser (<b>e</b>–<b>h</b>) or the sham groups (<b>i</b>–<b>l</b>), the annulus area displayed a continuous, lumen-sided endothelial layer regardless of the timepoint or group (<b>m</b>–<b>x</b>). vWF (red), vimentin (green), with DAPI counterstain (blue); scale bars = 100 µm.</p>
Full article ">Figure 7
<p>Analysis of degenerative calcification. Representative pairs of cross-section images and their details for all groups at t = 0 (ROS (<b>a</b>,<b>c</b>), laser (<b>e</b>,<b>g</b>), sham (<b>i</b>,<b>k</b>)) and at t = 168 d ((ROS (<b>b</b>,<b>d</b>), laser (<b>f</b>,<b>h</b>)<b>,</b> sham (<b>j</b>,<b>l</b>)). Von Kossa staining: calcification (brownish/black) measured with nuclear fast red counterstain (red); scale bars = 1000 µm. This analysis showed hardly any hint of calcification in the cusps at t = 0 (<b>m</b>) but a slight increase over time, especially in the ROS group, which achieved significant differences at t = 168 d (<b>n</b>). * <span class="html-italic">p</span> &lt; 0.05. On the contrary, the results of the semiquantitative analysis (von Kossa score: 0–5) displayed slight hints of calcification in the annulus areas in some of the animals at t = 0 and t = 168 d, regardless of the groups, with hardly any aggravation over time (<b>o</b>,<b>p</b>).</p>
Full article ">Figure 7 Cont.
<p>Analysis of degenerative calcification. Representative pairs of cross-section images and their details for all groups at t = 0 (ROS (<b>a</b>,<b>c</b>), laser (<b>e</b>,<b>g</b>), sham (<b>i</b>,<b>k</b>)) and at t = 168 d ((ROS (<b>b</b>,<b>d</b>), laser (<b>f</b>,<b>h</b>)<b>,</b> sham (<b>j</b>,<b>l</b>)). Von Kossa staining: calcification (brownish/black) measured with nuclear fast red counterstain (red); scale bars = 1000 µm. This analysis showed hardly any hint of calcification in the cusps at t = 0 (<b>m</b>) but a slight increase over time, especially in the ROS group, which achieved significant differences at t = 168 d (<b>n</b>). * <span class="html-italic">p</span> &lt; 0.05. On the contrary, the results of the semiquantitative analysis (von Kossa score: 0–5) displayed slight hints of calcification in the annulus areas in some of the animals at t = 0 and t = 168 d, regardless of the groups, with hardly any aggravation over time (<b>o</b>,<b>p</b>).</p>
Full article ">Figure 8
<p>Analysis of the mean transvalvular pressure gradient. The measurement of the mean transvalvular pressure gradient just before termination of each experiment (<b>a</b>) displayed no significant differences at t = 0 (<b>b</b>), but an increase in all the groups over time up to t = 168 d (<b>c</b>) with a significant difference between the ROS and laser group as well as the ROS and sham group. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
11 pages, 894 KiB  
Article
The Effect of Intravascular Laser Irradiation of Blood on Serum Biomarkers and Clinical Outcome in Knee Osteoarthritis Patients: A Double-Blind Randomized Control Trial
by Yu-Chi Su, Yu-Ping Shen, Chih-Ya Chang, Ke-Ting Pan, Shih-Ming Huang and Liang-Cheng Chen
Int. J. Mol. Sci. 2024, 25(24), 13608; https://doi.org/10.3390/ijms252413608 - 19 Dec 2024
Viewed by 235
Abstract
Knee osteoarthritis (OA) is a prevalent degenerative joint disease globally, causing pain, stiffness, and disability. Intravascular laser irradiation of blood (ILIB) has been used for chronic pain and musculoskeletal disease. However, evidence on the clinical benefits and serum biomarkers post-ILIB therapy in knee [...] Read more.
Knee osteoarthritis (OA) is a prevalent degenerative joint disease globally, causing pain, stiffness, and disability. Intravascular laser irradiation of blood (ILIB) has been used for chronic pain and musculoskeletal disease. However, evidence on the clinical benefits and serum biomarkers post-ILIB therapy in knee OA is insufficient. We designed a double-blind randomized controlled trial to evaluate the clinical and biological outcomes of ILIB therapy for knee OA. Seventeen patients with knee OA were randomly assigned to the ILIB and control groups. The outcomes included the Western Ontario and McMaster Universities Osteoarthritis (WOMAC) Scale, visual analog scale, and biomarker analysis of interleukin (IL)-6, IL-13, IL-1β, epidermal growth factor, macrophage inflammatory protein-1β, and eotaxin. The measurements were performed at baseline and three days, one month, and three months post-intervention. The ILIB group showed a significant improvement in the WOMAC-pain score at one month of follow-up than the control group. IL-1β levels reduced significantly on day three, one month, and three months, and IL-13 levels reduced on day three and three months during follow-up in the ILIB group. ILIB therapy reduced knee OA pain for one month and significantly reduced serum IL-1β and IL-13 levels, suggesting potential for pain management. Full article
(This article belongs to the Special Issue Osteoarthritis Biomarkers, Diagnosis and Treatments)
Show Figures

Figure 1

Figure 1
<p>Study flow diagram.</p>
Full article ">Figure 2
<p>Biomarker assessment of ILIB and control group during follow-up time point: (<b>a</b>) IL-1β (<b>b</b>) IL-6 (<b>c</b>) IL-13 (<b>d</b>) EGF (<b>e</b>) MIP-1β (<b>f</b>) eotaxin; statistical analysis was performed using independent t-test or Mann–Whitney U test for intergroup comparisons (*: <span class="html-italic">p</span>-value &lt; 0.05); Wilcoxon singled-rank test for intra-group comparisons to the baseline (#: <span class="html-italic">p</span> value &lt; 0.05).</p>
Full article ">
15 pages, 4248 KiB  
Article
The In Vitro Enhancement of Retinal Cell Viability via m6A and m5C RNA Methylation-Mediated Changes in the Levels of Heme Oxygenase (HO-1) and DNA Damage Repair Molecules Using a 50 Hz Sinusoidal Electromagnetic Field (EMF)
by Gabriela Betlej, Ewelina Bator, Anna Koziorowska, Marek Koziorowski and Iwona Rzeszutek
Int. J. Mol. Sci. 2024, 25(24), 13606; https://doi.org/10.3390/ijms252413606 - 19 Dec 2024
Viewed by 238
Abstract
Degenerative retinal diseases can lead to blindness if left untreated. At present, there are no curative therapies for retinal diseases. Therefore, effective treatment strategies for slowing the progression of retinal diseases and thus improving patients’ life standards are urgently needed. The present study [...] Read more.
Degenerative retinal diseases can lead to blindness if left untreated. At present, there are no curative therapies for retinal diseases. Therefore, effective treatment strategies for slowing the progression of retinal diseases and thus improving patients’ life standards are urgently needed. The present study aimed to assess the effect of sinusoidal electromagnetic field (EMF) (50 Hz, 1.3 mT) treatment for 15 and 30 min on spontaneously arising retinal pigment epithelial cells (ARPE-19) and retinal ganglion cells (RGC-5) and its short-term post-treatment significance. Our study indicated the beneficial impact of EMF treatment on the proliferative and migratory capacity of the tested cells. ARPE-19 and RGC-5 cells exposed to an EMF exhibited elevated levels of HO-1, increased N6-methyladenosine (m6A) and N5-methylcytosine (m5C) status mediated by METTL3 and NSUN2, respectively, and changes in levels of DNA damage repair factors, which may contribute to the regenerative properties of ARPE-19 and RGC-5 cells. Overall, this analysis showed that EMF (sinusoidal, 50 Hz, 1.3 mT) treatment may serve as a potential therapeutic strategy for retinal diseases. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Electromagnetic field (EMF)-mediated changes in the ARPE-19 and RGC-5 cells’ fitness. (<b>A</b>) Changes in the cell viability of ARPE-19 and RGC-5 cells upon stimulation with the EMF. Cells were treated with a sinusoidal EMF (50 Hz; 1.3 mT) for 15 and 30 min and cell viability was analyzed at 0, 1, 2, 3, 6, 12, 24, and 36 h post-treatment using a RealTime-Glo™ MT Cell Viability Assay. Data were normalized to the 0 h time point. The line graphs indicate SD, <span class="html-italic">n</span> = 3 for each time point, *** <span class="html-italic">p</span> &lt; 0.001, * <span class="html-italic">p</span> &lt; 0.05, compared to the corresponding untreated control (CTR) (ANOVA and Dunnett’s a posteriori test), <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 30 min of EMF treatment compared to 15 min of EMF treatment (one-way ANOVA and Tukey’s a posteriori test). (<b>B</b>) The levels of LDH were analyzed with a LDH-Glo™ Cytotoxicity Assay after 1, 6, 12, and 24 h of EMF treatment for 15 and 30 min and are presented as [mU/mL]. Bars indicate SD, <span class="html-italic">n</span> = 3, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 compared to the corresponding untreated control (CTR) (ANOVA and Dunnett’s a posteriori test), <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 30 min of treatment compared to 15 min of EMF treatment (one-way ANOVA and Tukey’s a posteriori test). (<b>C</b>,<b>D</b>) The efficacy of wound healing of ARPE-19 and RGC-5 cells was analyzed up to 24 h post-treatment with the EMF for 15 and 30 min. Bars indicate SD, <span class="html-italic">n</span> = 12, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 compared to the corresponding untreated control (CTR) (ANOVA and Dunnett’s a posteriori test), <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 30 min of treatment compared to 15 min of EMF treatment (one-way ANOVA and Tukey’s a posteriori test). Representative microphotographs are shown (<b>D</b>). Observations were made at 20× magnification. (<b>E</b>) Levels of Bcl-2 were detected using an immunostaining protocol and imaging cytometry. Results are presented as relative fluorescence units (RFU). Box and whisker plots are shown, <span class="html-italic">n</span> = 3, *** <span class="html-italic">p</span> &lt; 0.001, * <span class="html-italic">p</span> &lt; 0.05 compared to the corresponding untreated control (CTR) (ANOVA and Dunnett’s a posteriori test), <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, 30 min EMF treatment compared to 15 min of EMF treatment (one-way ANOVA and Tukey’s a posteriori test). CTR—control conditions. EMF-treated cells—cells treated with 15 and 30 min of a sinusoidal electromagnetic field with a frequency of 50 Hz and intensity of 1.3 mT.</p>
Full article ">Figure 2
<p>Electromagnetic field (EMF)-mediated changes in heme oxygenase 1 (HO-1) and heme oxygenase 2 (HO-2) levels in ARPE-19 and RGC-5 cells. Western blot-based analysis of HO-1 and HO-2 protein levels in CTR (untreated cells) and EMF-treated ARPE-19 and RGC-5 cells. The effect of EMF treatment on HO-1 and HO-2 protein levels was assessed at the 0 h, 24 h, and 48 h time points. Anti-GAPDH served as a loading control. Data were normalized to GAPDH. Bars indicate SD, <span class="html-italic">n</span> ≥ 2 ** <span class="html-italic">p</span> &lt; 0.01 compared to the corresponding untreated control (CTR) (ANOVA and Dunnett’s a posteriori test), <sup>^^</sup> <span class="html-italic">p</span> &lt; 0.01 compared to the corresponding 0 h time point (one-way ANOVA and Tukey’s a posteriori test). CTR—control conditions. EMF-treated cells—cells treated with 15 and 30 min of a sinusoidal electromagnetic field with a frequency of 50 Hz and intensity of 1.3 mT.</p>
Full article ">Figure 3
<p>Analysis of m<sup>6</sup>A and m<sup>5</sup>C RNA methylation status and protein levels of the RNA methyltransferases METTL3 and NSUN2 in CTR and EMF-treated ARPE-19 and RGC-5 cells. (<b>A</b>) ELISA-based assessment of RNA:m<sup>5</sup>C methylation levels in CTR and EMF-treated ARPE-19 and RGC-5 cells, presented as [%]. Analysis of RNA:m<sup>5</sup>C methylation levels in cells exposed to EMF treatment for 15 min and 30 min were made at the 0 h, 12 h, and 24 h time points. Untreated cells served as a control (CTR). Bars indicate SD, <span class="html-italic">n</span> = 2. (<b>B</b>) Analysis of RNA:m<sup>6</sup>A methylation levels [%] in CTR and EMF-treated ARPE-19 and RGC-5 cells using ELISA. Analysis of RNA:m<sup>6</sup>A methylation levels in cells exposed to EMF treatment for 15 min and 30 min were made at the 0 h, 12 h, and 24 h time points. Untreated cells served as a control (CTR). Bars indicate SD = 2. (<b>C</b>) Western blot-based analysis of protein levels of the METTL3 and NSUN2 methyltransferases in CTR and EMF-treated ARPE-19 and RGC-5 cells. The effect of EMF treatment on METTL3 and NSUN2 protein levels was measured at the 0 h, 24 h, and 48 h time points. The anti-GAPDH antibody served as a loading control. Data were normalized to GAPDH. Bars indicate SD, <span class="html-italic">n</span> ≥ 2, * <span class="html-italic">p</span> &lt; 0.05 compared to the corresponding untreated control (CTR) (ANOVA and Dunnett’s a posteriori test). CTR—control conditions (untreated cells). EMF-treated cells—cells treated with 15 and 30 min of a sinusoidal electromagnetic field with a frequency of 50 Hz and intensity of 1.3 mT.</p>
Full article ">Figure 4
<p>Electromagnetic field (EMF)-mediated DNA damage response in ARPE-19 and RGC-5 cells. (<b>A</b>) Immunostaining- and imaging cytometry-based 53BP1 levels. Results are presented as relative fluorescence units (RFU). Box and whisker plots are shown, <span class="html-italic">n</span> = 3, * <span class="html-italic">p</span> &lt; 0.05 compared to the corresponding untreated control (CTR) (ANOVA and Dunnett’s a posteriori test). (<b>B</b>) Western blot-based analysis of the levels of proteins involved in DNA damage repair in ARPE-19 and RGC-5 cells. Anti-GAPDH served as a loading control. Data were normalized to GAPDH. Bars indicate SD, <span class="html-italic">n</span> ≥ 2, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 compared to the corresponding untreated control (CTR) (ANOVA and Dunnett’s a posteriori test), <sup>^</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the corresponding 0 h time point (one-way ANOVA and Tukey’s a posteriori test). CTR—control conditions, untreated cells. EMF-treated cells—cells treated with 15 and 30 min of a sinusoidal electromagnetic field with a frequency of 50 Hz and intensity of 1.3 mT and analyzed at 0 h, 24 h, and 48 h post-treatment.</p>
Full article ">Figure 5
<p>Scheme showing the effect of the sinusoidal electromagnetic field (EMF) with an induction of 50 Hz and frequency of 1.3 mT on ARPE-19 and RGC-5 cells. Upward arrows (↑) indicate an up-regulation, while downward arrows (↓) indicate a down-regulation.</p>
Full article ">
14 pages, 7660 KiB  
Article
Boric Acid Protects the Uterus and Fallopian Tubes from Cyclophosphamide-Induced Toxicity in a Rat Model
by Enes Karaman and Adem Yavuz
Pharmaceuticals 2024, 17(12), 1716; https://doi.org/10.3390/ph17121716 - 19 Dec 2024
Viewed by 336
Abstract
Background/Objectives: Cyclophosphamide (CP) is widely used for treating various cancers and autoimmune diseases, but it causes damage to reproductive organs due to oxidative stress (OS) and inflammation. Boric acid (BA) has antioxidant properties that may help reduce OS, which is critical for [...] Read more.
Background/Objectives: Cyclophosphamide (CP) is widely used for treating various cancers and autoimmune diseases, but it causes damage to reproductive organs due to oxidative stress (OS) and inflammation. Boric acid (BA) has antioxidant properties that may help reduce OS, which is critical for preserving uterine functionality, particularly for cancer patients considering pregnancy after cryopreservation. This study aimed to determine whether BA could diminish CP-induced toxicity in the uterus and fallopian tubes (FT) using CP-induced toxicity in a rat model. Methods: Forty female Wistar rats, aged 18–20 weeks, were divided into four groups as follows: control, oral BA (OBR), CP, and CP plus OBR (CP + OBR). The toxicity was induced in the CP and CP + OBR groups with an initial dose of 200 mg/kg CP, followed by 8 mg/kg daily for 14 days. Rats in the OBR and CP + OBR groups received 20 mg/kg/day of BA. After the 16-day experiment, tissues were collected for analysis. Results: Histopathological and immunohistochemical assessments of IL-6 and HIF-1α expressions were used to evaluate inflammation and OS. The control, OBR, and CP + OBR groups maintained normal tissue features, while the CP group showed epithelial cell shedding, vacuolization, degenerative endometrial glands, lymphocyte infiltration, and reduced collagen fiber density. Elevated HIF-1α and IL-6 expressions in the uterus and FT indicated significant OS and inflammation. Conclusions: The study concluded that BA supplementation in CP-treated rats effectively reduced CP-induced uterine and FT damage, suggesting the potential protective role of BA in managing CP-associated toxicity. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The evaluation of histopathological parameters, shedding/vacuolization in epithelial cells, degenerative endometrial glands, lymphocyte infiltration, and collagen fiber of the uterus (<b>a</b>) and fallopian tube (<b>b</b>) across the control, OBR, CP, and CP + OBR groups. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Histopathological examination of the uterus by H&amp;E staining in control (<b>a</b>), OBR (<b>b</b>), CP (<b>c</b>), and CP + OBR (<b>d</b>) groups. Scale bars: 200 µm. Endometrial glands (◆), shedding epithelial cells (arrow), vacuolization in epithelial cells (arrowhead), lymphocyte infiltration areas (*).</p>
Full article ">Figure 3
<p>Histopathological examination of the uterus by MT staining in control (<b>a</b>), OBR (<b>b</b>), CP (<b>c</b>), and CP + OBR (<b>d</b>) groups. Scale bars: 200 µm. Endometrial glands (◆), shedding epithelial cells (arrow).</p>
Full article ">Figure 4
<p>Histopathological examination of the fallopian tube by H&amp;E staining in control (<b>a</b>), OBR (<b>b</b>), CP (<b>c</b>), and CP + OBR (<b>d</b>) groups. Scale bars: 200 µm. Shedding epithelial cells (arrow), vacuolization in epithelial cells (arrowhead).</p>
Full article ">Figure 5
<p>Histopathological examination of the fallopian tube by MT staining in control (<b>a</b>), OBR (<b>b</b>), CP (<b>c</b>), and CP + OBR (<b>d</b>) groups. Scale bars: 100 µm.</p>
Full article ">Figure 6
<p>The statistical analysis of HIF-1α (<b>a</b>,<b>c</b>) and IL-6 (<b>b</b>,<b>d</b>) immunoreactivity in the uterus and fallopian tubes from the control, OBR, CP, and CP + OBR groups. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001; ns, not significant/<span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>The immunoreactivity of HIF-1α in rat uterus from the control (<b>a</b>), OBR (<b>b</b>), CP (<b>c</b>), and CP + OBR (<b>d</b>) groups. Scale bars: 100 µm. Immunopositive cells (arrows).</p>
Full article ">Figure 8
<p>The immunoreactivity of IL-6 in rat uterus from the control (<b>a</b>), OBR (<b>b</b>), CP (<b>c</b>), and CP + OBR (<b>d</b>) groups. Scale bars: 100 µm. Immunopositive cells (arrows).</p>
Full article ">Figure 9
<p>The immunoreactivity of HIF-1α in rat fallopian tubes from the control (<b>a</b>), OBR (<b>b</b>), CP (<b>c</b>), and CP + OBR (<b>d</b>) groups. Scale bars: 100 µm. Immunopositive cells (arrows).</p>
Full article ">Figure 10
<p>The immunoreactivity of IL-6 in rat fallopian tubes from the control (<b>a</b>), OBR (<b>b</b>), CP (<b>c</b>), and CP + OBR (<b>d</b>) groups. Scale bars: 100 µm. Immunopositive cells (arrows).</p>
Full article ">
12 pages, 3039 KiB  
Review
Intervertebral Disc Degeneration and Regeneration: New Molecular Mechanisms and Therapeutics: Obstacles and Potential Breakthrough Technologies
by William Taylor and William Mark Erwin
Cells 2024, 13(24), 2103; https://doi.org/10.3390/cells13242103 - 19 Dec 2024
Viewed by 337
Abstract
Pain and disability secondary to degenerative disc disease continue to burden the healthcare system, creating an urgent need for effective, disease-modifying therapies. Contemporary research has identified potential therapies that include protein-, cellular- and/or matrix-related approaches; however, none have yet achieved a meaningful clinical [...] Read more.
Pain and disability secondary to degenerative disc disease continue to burden the healthcare system, creating an urgent need for effective, disease-modifying therapies. Contemporary research has identified potential therapies that include protein-, cellular- and/or matrix-related approaches; however, none have yet achieved a meaningful clinical impact. The tissue-specific realities of the intervertebral disc create considerable therapeutic challenges due to the disc’s location, compartmentalization, hypovascularization and delicate physiological environment. Furthermore, the imaging modalities currently used in practice are largely unable to accurately identify sources of pain ostensibly discogenic in origin. These obstacles are considerable; however, recent research has begun to shed light on possible breakthrough technologies. Such breakthroughs include revolutionary imaging to better identify tissue sources of pain. Furthermore, novel molecular therapies have been shown to be able to mediate the progression of degenerative disc disease in some large animal studies, and even provide some insight into suppressing the development of tissue sources of discogenic pain. These potential breakthrough technologies have yet to be translated for clinical use. Full article
Show Figures

Figure 1

Figure 1
<p>Images of non-chondrodystrophic and chondrodystrophic canine IVDs. (<b>a</b>) Non-chondrodystrophic canine IVD sourced from 3-year-old outbred canine. (<b>b</b>) 3-year-old chondrodystrophic canine (Beagle) IVD. Note the gelatinous appearance of the non-chondrodystrophic canine compared to the fibrocartilaginous appearance of the chondrodystrophic canine.</p>
Full article ">Figure 2
<p>Schematic of pro-inflammatory intracellular signaling induced by IL-1β, TNF-α, IL-6, IL-8 and Fas Ligand and the anti-inflammatory and pro-anabolic effects induced by the intra-discal injection of TGF-β1 and CTGF. The pro-inflammatory cytokines, principally IL-1β, induce downstream activation of P38 MAPK and translocation of the NFκβ complex to the nucleus that in turn increase inflammation (IL-6, IL-8, and TNF-α) and extracellular matrix remodeling enzymes such as MMP-3, -13, ADAMTS 4/5. The addition of TGF-β and CTGF suppresses the expression of these inflammatory proteins and suppresses cell death via suppression of the activation of Caspase-3.</p>
Full article ">
19 pages, 3101 KiB  
Review
Lower Limb Osteochondrosis and Apophysitis in Young Athletes—A Comprehensive Review
by Krystian Maruszczak, Tomasz Madej and Piotr Gawda
Appl. Sci. 2024, 14(24), 11795; https://doi.org/10.3390/app142411795 - 17 Dec 2024
Viewed by 326
Abstract
Osteochondrosis and apophysitis are common causes of pain in the growing skeleton, each with different causes and treatment approaches. Osteochondrosis involves degenerative processes affecting the epiphyseal ossification centers of the developing bones, while apophysitis results from repetitive traction injuries to the tendon insertions [...] Read more.
Osteochondrosis and apophysitis are common causes of pain in the growing skeleton, each with different causes and treatment approaches. Osteochondrosis involves degenerative processes affecting the epiphyseal ossification centers of the developing bones, while apophysitis results from repetitive traction injuries to the tendon insertions and affects both the cartilage and the underlying bone. Raising awareness of these conditions is crucial to facilitate the recovery of young athletes and prevent their sporting careers from being jeopardized early on. This review presents six known lower limb conditions that occur in young athletes, including three apophysitis such as Osgood–Schlatter disease, Sinding–Larsen–Johansson syndrome, and Sever’s disease, and three osteochondroses, including Perthes’ disease, Köhler’s disease, and Freiberg’s disease. The aim of this review is to outline the pathophysiology, clinical presentation, and treatment strategies for each of these conditions to provide a comprehensive understanding of their impact on young athletes. This review will provide clinicians, coaches, and physiotherapists with essential, evidence-based insights to increase their awareness of these conditions and refine treatment strategies for young athletes. Full article
(This article belongs to the Special Issue Advanced Image Analysis and Processing Technologies and Applications)
Show Figures

Figure 1

Figure 1
<p>Osgood–Schlatter disease on the ultrasound image. (<b>a</b>) Irregular contour of the ossification center of the tibial tuberosity apophysis. (<b>b</b>) Increased vascularity at the attachment site of the patellar ligament observed on Power Doppler ultrasound.</p>
Full article ">Figure 2
<p>Sinding–Larsen–Johansson syndrome on the ultrasound image. (<b>a</b>,<b>b</b>) Irregular contour of the patellar apex.</p>
Full article ">Figure 3
<p>Sever’s disease on the ultrasound image. Irregular contours of the cortical layer of the calcaneal tuberosity are noted. (<b>a</b>) Left calcaneal tuberosity. (<b>b</b>) Right calcaneal tuberosity.</p>
Full article ">Figure 4
<p>Legg–Calvé–Perthes disease on the ultrasound image. (<b>a</b>) Inflammatory changes with joint capsule vascularization on Power Doppler ultrasound. (<b>b</b>) Joint capsule thickening to 8 mm with decreased echogenicity. (<b>c</b>,<b>d</b>) Inflammatory changes in the joint with a thickened, hypoechoic joint capsule and a collapsed femoral head.</p>
Full article ">Figure 4 Cont.
<p>Legg–Calvé–Perthes disease on the ultrasound image. (<b>a</b>) Inflammatory changes with joint capsule vascularization on Power Doppler ultrasound. (<b>b</b>) Joint capsule thickening to 8 mm with decreased echogenicity. (<b>c</b>,<b>d</b>) Inflammatory changes in the joint with a thickened, hypoechoic joint capsule and a collapsed femoral head.</p>
Full article ">Figure 5
<p>Comparative ultrasound image showing the right side (R LONG) affected by the pathological process and the normal left side (L LONG) in a longitudinal section along the femoral neck.</p>
Full article ">Figure 6
<p>A summary of the locations of apophysitis and osteochondrosis.</p>
Full article ">
15 pages, 4209 KiB  
Article
Construction of Nicotinamide Mononucleotide-Loaded Liposomes and Their In Vitro Transport Across the Blood–Brain Barrier
by Tiantian Wang, Qi Wu, Lihong Wang, Tao Lan, Zhenyu Yun, Lin Zhao and Xi Wu
Appl. Sci. 2024, 14(24), 11732; https://doi.org/10.3390/app142411732 - 16 Dec 2024
Viewed by 491
Abstract
Nicotinamide mononucleotide (NMN) possesses a variety of physiological functions and has therapeutic effects on cardio-cerebral diseases, senile degenerative diseases, neurodegenerative diseases, delayed aging, etc. However, its ability to cross the blood–brain barrier (BBB) and the mechanism of its transport have not been reported. [...] Read more.
Nicotinamide mononucleotide (NMN) possesses a variety of physiological functions and has therapeutic effects on cardio-cerebral diseases, senile degenerative diseases, neurodegenerative diseases, delayed aging, etc. However, its ability to cross the blood–brain barrier (BBB) and the mechanism of its transport have not been reported. In this study, we used the immortalized hCMEC/D3 cell line to construct an in vitro monolayer cell BBB model, evaluated its ability to cross the blood–brain barrier, and explored the mechanism by carrying out transport and efflux experiments on NMN. The ability of NMN to cross the BBB was investigated by preparing NMN-loaded liposomes conjugated with ANG peptide and RVG peptide. The results showed that the transmembrane transport ability of NMN was moderate, and the transport mechanism was passive transport relying on the concentration difference. The trans-BBB ability of ANG peptide coupled with NMN could be highly significantly improved. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic diagram of targeted liposome synthesis.</p>
Full article ">Figure 2
<p>Liposome characterization: (<b>a</b>) blank liposomes (KB-lips); (<b>b</b>) results of liposome particle size.</p>
Full article ">Figure 3
<p>Time-dependent TEER values of monolayer BBB in vitro.</p>
Full article ">Figure 4
<p>MTT method detection results: (<b>a</b>) NMN; (<b>b</b>) KB-lips; (<b>c</b>) NMN-lips; (<b>d</b>) ANG-NMN-lips; (<b>e</b>) RVG-NMN-lips; *: compared with the blank group, there is a significant difference, <span class="html-italic">p</span> &lt; 0.05; **: compared with the blank group, there is a highly significant difference, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4 Cont.
<p>MTT method detection results: (<b>a</b>) NMN; (<b>b</b>) KB-lips; (<b>c</b>) NMN-lips; (<b>d</b>) ANG-NMN-lips; (<b>e</b>) RVG-NMN-lips; *: compared with the blank group, there is a significant difference, <span class="html-italic">p</span> &lt; 0.05; **: compared with the blank group, there is a highly significant difference, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>The transport kinetics curve and transport percentage in NMN time-dependency bidirectional transmembrane transport experiment and the MTT method detection results: (<b>a</b>) transport kinetics curve of AP-BL; (<b>b</b>) transport kinetics curve of BL-AP; (<b>c</b>) transport percentage of AP-BL; (<b>d</b>) transport percentage BL-AP.</p>
Full article ">Figure 6
<p>The transport kinetics curve and transport percentage in the NMN concentration-dependency bidirectional transmembrane transport experiment: (<b>a</b>) transport kinetics curve of AP-BL; (<b>b</b>) transport kinetics curve of BL-AP; (<b>c</b>) transport percentage of AP-BL; (<b>d</b>) transport percentage BL-AP.</p>
Full article ">Figure 6 Cont.
<p>The transport kinetics curve and transport percentage in the NMN concentration-dependency bidirectional transmembrane transport experiment: (<b>a</b>) transport kinetics curve of AP-BL; (<b>b</b>) transport kinetics curve of BL-AP; (<b>c</b>) transport percentage of AP-BL; (<b>d</b>) transport percentage BL-AP.</p>
Full article ">Figure 7
<p>Results of four types of liposome transmembrane transport: (<b>a</b>) AP-BL; (<b>b</b>) BL-AP. #: compared with the NMN group, there is a significant difference, <span class="html-italic">p</span> &lt; 0.05; ##: compared with the NMN group, there is a highly significant difference, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
20 pages, 3210 KiB  
Article
Inhibiting De Novo Biosynthesis of Ceramide by L-Cycloserine Can Prevent Light-Induced Retinal Degeneration in Albino BALB/c Mice
by Faiza Tahia, Dejian Ma, Daniel J. Stephenson, Sandip K. Basu, Nobel A. Del Mar, Nataliya Lenchik, Harry Kochat, Kennard Brown, Charles E. Chalfant and Nawajes Mandal
Int. J. Mol. Sci. 2024, 25(24), 13389; https://doi.org/10.3390/ijms252413389 - 13 Dec 2024
Viewed by 475
Abstract
Retinal degenerative diseases lead to irreversible vision loss due to photoreceptor cell death, driven by complex genetic and environmental factors. Ceramide, a sphingolipid metabolite, emerges as a critical mediator in the apoptotic cascade associated with retinal degeneration. Our previous work demonstrated L-Cycloserine’s ability [...] Read more.
Retinal degenerative diseases lead to irreversible vision loss due to photoreceptor cell death, driven by complex genetic and environmental factors. Ceramide, a sphingolipid metabolite, emerges as a critical mediator in the apoptotic cascade associated with retinal degeneration. Our previous work demonstrated L-Cycloserine’s ability to protect photoreceptor-derived cells from oxidative stress by inhibiting the de novo ceramide pathway and thus prompting further investigation on its effect in the in vivo retina. This study investigates the potential of L-Cycloserine to protect albino BALB/c mice against light-induced retinal degeneration (LIRD). L-Cycloserine, in an optimal dose, administered systemically 30 min before LIRD, was found to prevent photoreceptor cell death significantly from light-induced degeneration. We further determined the retinal bioavailability and pharmacokinetic behavior of L-Cycloserine, its effect on sphingolipid profile, expression of sphingolipid biosynthetic, and cell death-promoting genes and proteins from the retina to understand the underlying mechanisms. This study lays the groundwork for further preclinical and clinical investigations into L-Cycloserine’s potential as a novel therapeutic in treating retinal degenerative diseases. Full article
Show Figures

Figure 1

Figure 1
<p>L-Cycloserine protects mouse retina from LIRD. ERG analysis shows Scotopic a-wave (<b>A</b>), b-wave (<b>B</b>), and Photopic b-wave (<b>C</b>) from NLD: no light-damaged control group; VLD: vehicle-injected light-damaged group; L-Cs 10 LD: L-Cycloserine-treated (10 mg/kg) light-damaged group. (n = 6/group; values are mean ± SEM; * represents significance between VLD and L-Cs 10 LD; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, by the student <span class="html-italic">t</span> test; # represents significance between NLD and VLD; # <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, by the student <span class="html-italic">t</span> test).</p>
Full article ">Figure 2
<p>A 10 mg/kg dose of L-Cycloserine provides maximum protection of mouse retina from LIRD. ERG analysis shows Scotopic a-wave (<b>A</b>), b-wave (<b>B</b>), and Photopic b-wave (<b>C</b>) from NLD: no light-damaged control group; VLD: vehicle-injected light-damaged group; L-Cs 5 LD,10 LD, 20 LD, and 40 LD: L-Cycloserine-treated (5, 10, 20, and 40 mg/kg, respectively) light-damaged group. n = 13 (NLD, VLD); n = 9 (L-Cs 5 LD, L-Cs 10 LD); n = 10 (L-Cs 20 LD); n = 8 (L-Cs 40 LD); values are mean ± SEM; * represents significance between VLD and L-Cycloserine treated group; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, by the student <span class="html-italic">t</span> test; # represents significance between NLD and VLD; # <span class="html-italic">p</span> &lt; 0.05, ### <span class="html-italic">p</span> &lt; 0.001, by the student <span class="html-italic">t</span> test).</p>
Full article ">Figure 3
<p>A 10 mg/kg dose of L-Cycloserine provides maximum protection of mouse retina from LIRD. Representative retinal histological sections from each treatment: (<b>A</b>): NLD, no light-damaged control; (<b>B</b>): VLD, vehicle-injected light-damaged group; (<b>C</b>–<b>F</b>): L-Cs 5 LD,10 LD, 20 LD, and 40 LD, represents L-Cycloserine-treated (5, 10, 20, and 40 mg/kg, respectively) light-damaged group. Scale bar in F represents 50 microns. (<b>G</b>) Quantitative morphometric measurement of ONL nuclei count from H and E-stained slides. n = 12 (NLD); n = 16 (VLD); n = 10 (L-Cs 5 LD); n = 13 (L-Cs 10 LD); n = 14 (L-Cs 20 LD); n = 11 (L-Cs 40 LD). Values are mean ± SEM; * represents significance between VLD and L-Cycloserine treated group; * <span class="html-italic">p</span> &lt; 0.001, by the student <span class="html-italic">t</span> test; # represents significance between NLD and VLD; # <span class="html-italic">p</span> &lt; 0.001, by the student <span class="html-italic">t</span> test. Abbreviations for retinal section: RPE, retinal pigment epithelium; PR, photoreceptors; OS, outer segments; IS, inner segments; ONL, outer nuclear layer; OPL, outer plexiform layer; INL, inner nuclear layer; IPL, inner plexiform layer; GCL, ganglion cell layer.</p>
Full article ">Figure 4
<p>Pharmacokinetic profile of L-Cycloserine in plasma and various tissues. L-cycloserine was intraperitoneally injected at a dose of 20 mg/kg and quantified using LC-MS/MS in retina (<b>A</b>), plasma (<b>B</b>), liver (<b>C</b>), and brain (<b>D</b>) at various time points after administration. The results are expressed as a mean ± SEM (n = 7 for each time point).</p>
Full article ">Figure 5
<p>Light damage and L-Cycloserine treatment alter ceramide and other sphingolipid levels in light-damaged BALB/c mice retina. (<b>A</b>–<b>F</b>): Analysis of major sphingolipid levels by absolute value (pmol/mg of tissue) showing the total composition of ceramide (Cer), sphingomyelin (SM), and monohexosylceramide (MHC) percent in BALB/c mice retina (n = 10/group). Analysis of selected ceramide species by absolute value (pmol/mg of tissue) in BALB/c mice retina at 0 h and 6 h after light damage (<b>G</b>–<b>J</b>). (n = 10/group, values are mean ± SEM; # represents significance between NLD and VLD; # <span class="html-italic">p</span> &lt; 0.05, ### <span class="html-italic">p</span> &lt; 0.001, by student <span class="html-italic">t</span>-test). NLD: no light-damaged control group, VLD 0: vehicle-injected light-damaged group at 0 h after light damage, L-Cs 0: L-Cycloserine (10 mg/kg) treated light-damaged group at 0 h after light damage, VLD 6: vehicle-injected light-damaged group at 6 h after light damage and L-Cs 6: L-Cycloserine (10 mg/kg) treated light-damaged group at 6 h after light damage.</p>
Full article ">Figure 6
<p>L-Cycloserine modulates retinal gene expression in light-damaged BALB/c mice. Quantitative analysis of gene expression of no light-damaged control (NLD), vehicle-injected light-damaged (VLD), and L-Cycloserine (10 mg/kg) treated light-damaged (L-Cs 10 LD) retina 0 h after light damage. Expression values (±SEM) are presented against fold change over control value (NLD), which was set to 1.0 (n = 6; # represents significance between NLD and VLD; # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, by the student <span class="html-italic">t</span> test; * represents significance between VLD and L-Cs 10 LD; * <span class="html-italic">p</span> &lt; 0.05, by the student <span class="html-italic">t</span> test).</p>
Full article ">Figure 7
<p>L-Cycloserine modulates expression of retinal proteins in light-damaged BALB/c mice. (<b>A</b>): Expression and quantification of heme oxygenase 1 (Ho1), Poly (ADP-ribose) polymerase (PARP) and Cathepsin D in BALB/c retina from no light-damaged control (NLD), vehicle-injected light-damaged (VLD) and L-Cycloserine (10 mg/kg) treated light-damaged (L-Cs 10 LD) group, 0 h after light damage. Quantification of (<b>B</b>): Ho1 (<b>C</b>) PARP (<b>D</b>): Cathepsin D in retinal tissue obtained with densitometric analysis and normalized with β-actin. (n = 3; # represents significance between NLD and VLD; ### <span class="html-italic">p</span> &lt; 0.001, by student <span class="html-italic">t</span>-test; * represents significance between VLD and L-Cs 10 LD; *** <span class="html-italic">p</span> &lt; 0.001, by student <span class="html-italic">t</span>-test).</p>
Full article ">
20 pages, 1333 KiB  
Review
Innovative Biotherapies and Nanotechnology in Osteoarthritis: Advancements in Inflammation Control and Cartilage Regeneration
by Binhan Liu, Tao Liu, Yanhong Li and Chunyu Tan
Int. J. Mol. Sci. 2024, 25(24), 13384; https://doi.org/10.3390/ijms252413384 - 13 Dec 2024
Viewed by 411
Abstract
Osteoarthritis (OA) is among the most prevalent degenerative joint disorders worldwide, particularly affecting the aging population and imposing significant disability and economic burdens. The disease is characterized by progressive degradation of articular cartilage and chronic inflammation, with no effective long-term treatments currently available [...] Read more.
Osteoarthritis (OA) is among the most prevalent degenerative joint disorders worldwide, particularly affecting the aging population and imposing significant disability and economic burdens. The disease is characterized by progressive degradation of articular cartilage and chronic inflammation, with no effective long-term treatments currently available to address the underlying causes of its progression. Conventional therapies primarily manage symptoms such as pain and inflammation but fail to repair damaged tissues. Emerging biotherapies and regenerative medicine approaches offer promising alternatives by addressing cartilage repair and inflammation control at the molecular level. This review explores the recent advancements in biotherapeutic strategies, including mesenchymal stem cell (MSC) therapy, growth factors, and tissue engineering, which hold the potential for promoting cartilage regeneration and modulating the inflammatory microenvironment. Additionally, the integration of nanotechnology has opened new avenues for targeted drug delivery systems and the development of innovative nanomaterials that can further enhance the efficacy of biotherapies by precisely targeting inflammation and cartilage damage. This article concludes by discussing the current clinical applications, the ongoing clinical trials, and the future research directions necessary to confirm the safety and efficacy of these advanced therapies for OA management. With these advancements, biotherapies combined with nanotechnology may revolutionize the future of OA treatment by offering precise and effective solutions for long-term disease management and improved patient outcomes. Full article
(This article belongs to the Section Molecular Nanoscience)
Show Figures

Figure 1

Figure 1
<p>Clinical manifestations of OA.</p>
Full article ">Figure 2
<p>Biological treatment of OA. Biological therapy for OA refers to the use of biologics or bioengineering methods to treat OA. These therapies can include biological products such as biological agents, growth factors, stem cell therapy, gene therapy, and others.</p>
Full article ">Figure 3
<p>Application of nanotechnology in OA.</p>
Full article ">
12 pages, 1173 KiB  
Article
Kappa Free Light Chains in Multiple Sclerosis as a Marker of Intrathecal Humoral Response: A Sex-Disaggregated Study
by Raffaella Candeloro, Maila Galloppa, Laura Lombardo, Michele Laudisi, Sara Ghisellini, Giovanna Negri, Caterina Ferri, Carla Marcialis, Tiziana Bellini, Maura Pugliatti and Massimiliano Castellazzi
Diagnostics 2024, 14(24), 2798; https://doi.org/10.3390/diagnostics14242798 - 12 Dec 2024
Viewed by 492
Abstract
Background: Kappa free light chains (KFLCs) are emerging as promising biomarkers for intrathecal B cell activity for diagnosing multiple sclerosis (MS) through cerebrospinal fluid (CSF) analysis. In this study, we evaluated the ability of KFLC formulas to identify the presence of MS and [...] Read more.
Background: Kappa free light chains (KFLCs) are emerging as promising biomarkers for intrathecal B cell activity for diagnosing multiple sclerosis (MS) through cerebrospinal fluid (CSF) analysis. In this study, we evaluated the ability of KFLC formulas to identify the presence of MS and their agreement with the ‘gold standard’ of CSF IgG oligoclonal bands (OCBs). Methods: A total of 233 patients were included in this study: 149, comprising 43 males and 106 females, had MS, and the remainder, 40 males and 44 females, had other neurological diseases (ONDs). We evaluated the potential of KFLCs in terms of sensitivity, specificity, and accordance. All analyses were conducted using a sex-disaggregated approach. Results: KFLCs showed a high sensitivity for both sexes with respect to the diagnosis of MS, with values between 74.42% and 93.03%. The specificity of the various formulas was much lower for females when compared to males, with values between 45.45% and 59.09%, with a significant difference between the two sexes for the K Index > 5.9 (p = 0.0451). Cohen’s kappa showed substantial agreement for men and moderate agreement for women between the KFLC indices and OCB. Conclusions: This study highlights the potential of KFLCs as a biomarker for MS but emphasises the need for sex-specific thresholds to improve diagnostic accuracy. Full article
(This article belongs to the Special Issue Biomarkers: Diagnostic Indicators for Human Ailments)
Show Figures

Figure 1

Figure 1
<p>The ability of different indices of free kappa light chains (KFLC) to identify the diagnosis of multiple sclerosis was assessed. Sensitivity (<b>A</b>), specificity (<b>B</b>), and agreement (<b>C</b>) were evaluated for each parameter in the sex-disaggregated population. Abbreviations: URL—Uniform Resource Locator; fair agreement—range from 0.21 to 0.40; moderate agreement—range from 0.41 to 0.60; substantial agreement—range from 0.61 to 0.80. Fisher’s exact test was used to compared sensibility and specificity between males and females. *: <span class="html-italic">p</span> = 0.0451.</p>
Full article ">Figure 2
<p>The ability of different indices of free kappa light chains (KFLC) to identify the presence of intrathecal immunoglobulin synthesis was assessed using cerebrospinal-fluid-restricted oligoclonal IgG bands as the reference “gold standard”. Sensitivity (<b>A</b>), specificity (<b>B</b>), and agreement (<b>C</b>) were evaluated for each parameter in the sex-disaggregated population. Abbreviations: URL: Uniform Resource Locator; fair agreement: range from 0.21 to 0.40; moderate agreement: range from 0.41 to 0.60; substantial agreement: range from 0.61 to 0.80. Fisher’s exact test was used to compared sensibility and specificity between males and females: all <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">
Back to TopTop