[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
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,319)

Search Parameters:
Keywords = dexamethasone

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 865 KiB  
Article
Chronic Dexamethasone Disturbs the Circadian Rhythm of Melatonin and Clock Genes in Goats
by Liuping Cai, Qu Chen, Canfeng Hua, Liqiong Niu, Qijun Kong, Lei Wu and Yingdong Ni
Animals 2025, 15(1), 115; https://doi.org/10.3390/ani15010115 (registering DOI) - 6 Jan 2025
Abstract
Dex is a drug commonly used as an immunosuppressive and anti-inflammatory agent in humans and animals. GCs have a profound impact on melatonin expression and biological rhythm. However, the effect of chronic exposure to Dex on melatonin secretion and biological clock gene expression [...] Read more.
Dex is a drug commonly used as an immunosuppressive and anti-inflammatory agent in humans and animals. GCs have a profound impact on melatonin expression and biological rhythm. However, the effect of chronic exposure to Dex on melatonin secretion and biological clock gene expression in ruminants is still unclear. Ten goats were randomly divided into two groups: the control group was injected with saline, and the Dex-treated group was intramuscularly injected daily for 21 d with 0.2 mg/kg Dex. The rhythm of melatonin secretion in the plasma was disturbed in the Dex group, and the plasma and colon levels of melatonin were lower in the Dex group compared to the control group (p < 0.05). Dex leads to a significant decrease in the expression of Arylalkylamine N-acetyltransferase (AANAT), a key melatonin synthase, in the pineal gland and colon. Detecting intestinal leakage-related indices showed that diamine oxidase (DAO) and lipopolysaccharide (LPS) content increased significantly in the Dex group (p < 0.05). We also detected genes associated with biological rhythms in the plasma. In the control group, the five tested genes showed circadian rhythms, but the circadian rhythms of Clock, Cry1, Cry2, and Per2 were abolished or blunted by the Dex (p < 0.05). Protein levels of CLOCK and BMAL1 in the colon changed significantly (p < 0.05). In conclusion, the above experimental results show that chronic exposure to Dex leads to the disorder of the circadian rhythms of melatonin secretion and clock genes. Full article
(This article belongs to the Section Animal Physiology)
22 pages, 2493 KiB  
Article
Hydrogels of Poly(2-hydroxyethyl methacrylate) and Poly(N,N-dimethylacrylamide) Interpenetrating Polymer Networks as Dermal Delivery Systems for Dexamethasone
by Marin Simeonov, Bistra Kostova, Rositsa Mihaylova and Elena Vassileva
Pharmaceutics 2025, 17(1), 62; https://doi.org/10.3390/pharmaceutics17010062 (registering DOI) - 5 Jan 2025
Viewed by 220
Abstract
Background/Objectives: This study is an attempt to reveal the potential of two types of interpenetrating polymer network (IPN) hydrogels based on poly(2-hydroxyethyl methacrylate) (PHEMA) and poly(N,N-dimethylacrylamide) (PDMAM). These IPNs were evaluated for their potential for dermal delivery of the hydrophobic drug dexamethasone [...] Read more.
Background/Objectives: This study is an attempt to reveal the potential of two types of interpenetrating polymer network (IPN) hydrogels based on poly(2-hydroxyethyl methacrylate) (PHEMA) and poly(N,N-dimethylacrylamide) (PDMAM). These IPNs were evaluated for their potential for dermal delivery of the hydrophobic drug dexamethasone (DEX). Methods: The two types of IPNs were analyzed for their rheological behavior, swelling characteristics, and drug-loading capacity with DEX. Drug release profiles were studied in Franz diffusion cells in PBS media. Finally, the cytotoxicity of the PHEMA/PDMAM-based IPNs was studied against T-cell lymphoma cells (HUT-78) and a normal murine fibroblast cell line (CCL-1). Results: The rheological properties of these hydrogels show suitable mechanical properties for dermal application, with G′ values of ~10 kPa. From the rheological data, the mesh size of these hydrogels was found to be influenced by the type of the IPN and its composition, varying between 6.5 and 50 nm. The loading capacity of both IPN types and DEX entrapment efficiency were highly influenced by the IPN’s composition. The loading capacity of the IPNs can reach ~3.5%, with a DEX entrapment efficiency of ~35%. The PHEMA/PDMAM IPNs demonstrate an extended release profile with up to ~95% DEX released in 24 h, while PDMAM/PHEMA IPNs release no more than ~25% DEX in 24 h. The drug release profiles follow either non-Fickian diffusion (n~0.6) or case-II transport (n~0.9–1), depending on the IPN’s composition. The PHEMA/PDMAM-based materials were found to be non-cytotoxic against HUT-78 and CCL-1 cells. Conclusions: The study reveals that the IPNs of PHEMA and PDMAM appear to be suitable platforms for dermal delivery of dexamethasone as they have appropriate mechanical properties, providing tools to control drug loading and release, and they are biocompatible with human skin cells. Full article
(This article belongs to the Special Issue Therapeutic Approaches for Wound-Associated Skin Diseases)
Show Figures

Figure 1

Figure 1
<p>Storage modulus (G′) (full symbols) and loss modulus (G″) (empty symbols) for the straight PDMAM/PHEMA IPNs (<b>A</b>) and for the reverse PHEMA/PDMAM IPNs (<b>B</b>) obtained via frequency sweep experiments.</p>
Full article ">Figure 2
<p>Mesh sizes (ξ) of (<b>A</b>) PDMAM/PHEMA IPN and (<b>B</b>) PHEMA/PDMAM IPN hydrogels as a function of PDMAM content.</p>
Full article ">Figure 3
<p>ESR for PDMAM/PHEMA IPNs synthesized using PDMAM SNs with 0.1 mol.% PEGDA in water and in ethanol (EtOH) (<b>A</b>) and ESR for PHEMA/PDMAM IPNs in EtOH (<b>B</b>).</p>
Full article ">Figure 4
<p>ESR of PDMAM SN with 0.4 mol.% PEGDA (D04) and its IPN with PHEMA (D04H500), in water and in ethanol (EtOH).</p>
Full article ">Figure 5
<p>SEM images of fractured surfaces of PDMAM SNs and PDMAM/PHEMA IPNs. Red arrows point at the second phase domains of PHEMA.</p>
Full article ">Figure 6
<p>Dependence of DEX EE (<b>A</b>) and DL (<b>B</b>) in PHEMA SN and PHEMA/PDMAM IPNs as a function of their composition (<math display="inline"><semantics> <mrow> <msubsup> <mi mathvariant="normal">r</mi> <mi mathvariant="normal">D</mi> <mi mathvariant="normal">r</mi> </msubsup> </mrow> </semantics></math>).</p>
Full article ">Figure 7
<p>Dependence of DEX EE (<b>A</b>) and DL (<b>B</b>) in PDMAM SNs and PDMAM/PHEMA IPNs, synthesized using PDMAM SNs with 0.1 mol.% PEGDA as a function of their composition (<math display="inline"><semantics> <mrow> <msubsup> <mi mathvariant="normal">r</mi> <mi mathvariant="normal">D</mi> <mi mathvariant="normal">s</mi> </msubsup> </mrow> </semantics></math>).</p>
Full article ">Figure 8
<p>Drug release profiles of DEX from PHEMA SN (H1) and PHEMA/PDMAM IPNs (H250 and H500) (<b>A</b>) and from PDMAM SN (D01) and PDMAM/PHEMA IPNs (D01H125 and D01H500) (<b>B</b>).</p>
Full article ">Figure 9
<p>Cell viability of cutaneous T-cell lymphoma cells (HUT-78) and normal murine fibroblast cell line (CCL-1) following 72 h exposure to different concentrations of H1 and H500 PHEMA/PDMAM IPNs. All experiments were run in triplicate and data are expressed as the mean ± SD. Statistical significance of the data was assessed using a one-way ANOVA (<span class="html-italic">p</span>-value ≤ 0.05 was considered statistically significant).</p>
Full article ">
15 pages, 1101 KiB  
Article
Analysis of Glucocorticoids as Potential Adulterants in Cosmetic Products: A Dual Approach for Qualitative and Quantitative Evaluation Based on ELISA and HPLC-MS Methods
by Seyedeh Rojin Shariati Pour, Afsaneh Emamiamin, Martina Zangheri, Donato Calabria, Massimo Guardigli, Emanuele Porru, Jessica Fiori and Mara Mirasoli
Appl. Sci. 2025, 15(1), 414; https://doi.org/10.3390/app15010414 (registering DOI) - 4 Jan 2025
Viewed by 558
Abstract
The analysis of cosmetic products represents an important field of analytical chemistry, since the demand for new formulations is continuously increasing. Regulations about prohibited/regulated compounds are applied in each country. Among the substances that are banned in cosmetics, corticosteroids represent a potential harm [...] Read more.
The analysis of cosmetic products represents an important field of analytical chemistry, since the demand for new formulations is continuously increasing. Regulations about prohibited/regulated compounds are applied in each country. Among the substances that are banned in cosmetics, corticosteroids represent a potential harm for consumers since the prolonged exposure to these compounds can affect health status. However, corticosteroids can be found in cosmetics as an illegal addition since they are able to alleviate the symptoms of inflammatory skin problems. In this work, two different approaches for detecting corticosteroids as potential adulterants in cosmetic products were compared. First, a reversed-phase HPLC-MS method was optimized and fully validated in order to identify and quantify eight corticosteroids (methylprednisolone, beclomethasone, flunisolide, budesonide, betamethasone 17-valerate, beclomethasone dipropionate, flumethasone, and dexamethasone). This reference method was then compared with an enzyme-linked immunosorbent assay (ELISA). Indeed, immunological techniques allow for rapid, low-cost, and sensitive detection of target analytes even in complex matrices, and they can be performed with simple instrumentation and by non-skilled personnel. The application of these methods on spiked cosmetic products was compared in terms of performance and advantages in order to evaluate the possibility of exploiting a complementary approach for optimizing the time for and costs of the analysis. Full article
(This article belongs to the Special Issue Analytical Chemistry: Techniques and Applications)
Show Figures

Figure 1

Figure 1
<p>Corticosteroid chemical structures.</p>
Full article ">Figure 2
<p>Calibration curves obtained in PBS/THF (circle symbols), cream A (triangular symbols), and serum (square symbols) for the different corticosteroids.</p>
Full article ">
27 pages, 3818 KiB  
Article
In Situ Gelling Dexamethasone Oromucosal Formulation: Physical Characteristics Influencing Drug Delivery
by Daniel Krchňák, Ľudmila Balážová, Michal Hanko, Dominika Žigrayová and Miroslava Špaglová
Gels 2025, 11(1), 26; https://doi.org/10.3390/gels11010026 - 2 Jan 2025
Viewed by 233
Abstract
The study focuses on the development of an in situ gelling dexamethasone (DEX) oromucosal formulation designed for the treatment of aphthous stomatitis. Three series of formulations were prepared; a first series containing DEX suspended, a second series containing DEX and, in addition, mint [...] Read more.
The study focuses on the development of an in situ gelling dexamethasone (DEX) oromucosal formulation designed for the treatment of aphthous stomatitis. Three series of formulations were prepared; a first series containing DEX suspended, a second series containing DEX and, in addition, mint essential oil (EO), and a third series containing EO and DEX solubilized in propylene glycol (PG). In the composition, polymers in the role of mucoadhesive agent were interchanged (hydroxypropyl methylcellulose (HPMC), hydroxypropyl cellulose (HPC), hydroxyethyl cellulose (HEC), methyl cellulose (MC), carboxymethyl cellulose (CMC), and sodium carboxymethyl cellulose (NaCMC). Specifically, DEX was incorporated at a concentration of 0.1% (w/w) in each formulation. The influence of mint EO and DEX solubilization on the physical properties (pH measurements, rheological analysis, swelling ability, and texture analysis) and in vitro drug release was studied. Key findings revealed that HPMC-based formulation containing mint EO and PG exhibited best swelling properties (700 ± 46% after 5 h), adequate adhesiveness and in vitro drug release (34.7 ± 5.9%). Furthermore, the irritation potential assessed via the hen’s egg test on the chorioallantoic membrane (HET-CAM) demonstrated low irritancy risk. Finally, Fourier-transform infrared spectroscopy (FT-IR) showed no incompatibility between DEX and excipients. Overall, the research highlights the potential of mucoadhesive systems in improving the therapeutic efficacy of oromucosal drug delivery for managing painful oral lesions. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The comparison of the swelling ability of the formulations without essential oil (EO) (first series) and with EO and DEX solubilized in propylene glycol (PG) (third series). The difference in swelling ability of all formulations is statistically significant (*) compared to the reference samples A1 or A3 with HPMC.</p>
Full article ">Figure 2
<p>Macroscopic observation of changes in the sample formulations after 5 h of swelling in the artificial saliva.</p>
Full article ">Figure 3
<p>FT-IR spectra of DEX (red), formulation A3 with DEX (green), zoomed spectra of formulation A3 with DEX (blue), and formulation A3 without DEX (black).</p>
Full article ">Figure 4
<p>In vitro release profiles of DEX from the formulations without essential oil (EO), with essential oil (EO), and with essential oil (EO) and DEX solubilized in propylene glycol (PG). A1, A2 and A3 were used as the reference samples in the series of formulations being compared. NS indicates a non-significant difference, two asterisks (**) a significant difference at a high level with <span class="html-italic">p</span> ≤ 0.01, and three asterisks (***) a significant difference at a very high level with <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 5
<p>The amount of DEX (%) released after 4 h during in vitro release study from the formulations. The asterisk (*) indicates a significant difference with respect to the corresponding reference sample from series one, without EO and PG. The formulations without EO (blue), with EO (green), with EO and PG (yellow).</p>
Full article ">Figure 6
<p>The microscopic observation of the chorioallantoic membrane after application of the examined formulations (green circles—hemorrhage, yellow circles—vasodilatation).</p>
Full article ">
11 pages, 666 KiB  
Review
The Role of Corticosteroids in Non-Bacterial and Secondary Encephalitis
by Giusy Di Flumeri, Luca Gregorio Giaccari, Maria Caterina Pace, Maria Beatrice Passavanti, Vincenzo Pota, Vincenzo Riccardi, Simona Brunetti, Pasquale Sansone, Francesco Coppolino and Caterina Aurilio
Life 2024, 14(12), 1699; https://doi.org/10.3390/life14121699 - 22 Dec 2024
Viewed by 406
Abstract
Encephalitis affects 1.9 to 14.3 people per 100,000 each year, and the mortality rate varies but can be up to 40%. After the identification of a particular microorganism in a patient with encephalitis, appropriate antimicrobial therapy should be initiated. Corticosteroid therapy represents a [...] Read more.
Encephalitis affects 1.9 to 14.3 people per 100,000 each year, and the mortality rate varies but can be up to 40%. After the identification of a particular microorganism in a patient with encephalitis, appropriate antimicrobial therapy should be initiated. Corticosteroid therapy represents a therapeutic option in the treatment of primary central nervous system diseases due to its ability to reduce the inflammatory commitment of CNS and consequently reduce mortality rates regardless of the causative agent of injury. Corticosteroid therapy represents a therapeutic option in the treatment of primary central nervous system diseases. Their use is also recommended in meningitis with autoimmune etiology. While corticosteroids have repeatedly been used as adjunctive treatment in encephalitis of viral etiology, the scientific evidence supporting their effectiveness remains scarce. The use of standard doses recommended by the guidelines seems reasonable as an initial setting, especially when a definitive diagnosis of the causal agent is still awaited. The subsequent adjustment should be personalized based on the individual clinical response. Full article
(This article belongs to the Section Pharmaceutical Science)
Show Figures

Figure 1

Figure 1
<p>Cascade of inflammatory mediators. Illustration of the inflammatory response produced by oligodendrocytes, astrocytes, and microglia to viral infection. (ATP, adenosine triphosphate; DAergic, dopaminergic; ICAM1, intercellular adhesion molecule 1; MHCI, major histocompatibility complex class I; NF, necrosis factor; ROS, reactive oxygen species; SASP, senescence-associated secretory phenotype; VCAM, vascular cell adhesion molecule).</p>
Full article ">
8 pages, 3259 KiB  
Case Report
SARS-CoV-2 Infection of the Central Nervous System: A Case Report
by Trifon Valkov, Radka Argirova and George Dimitrov
Viruses 2024, 16(12), 1962; https://doi.org/10.3390/v16121962 - 21 Dec 2024
Viewed by 513
Abstract
Central nervous system (CNS) infections caused by SARS-CoV-2 are uncommon. This case report describes the clinical progression of a 92-year-old female who developed a persistent neuroinfection associated with SARS-CoV-2. The patient initially presented with progressive fatigue, catarrhal symptoms, and a fever (38.6 °C). [...] Read more.
Central nervous system (CNS) infections caused by SARS-CoV-2 are uncommon. This case report describes the clinical progression of a 92-year-old female who developed a persistent neuroinfection associated with SARS-CoV-2. The patient initially presented with progressive fatigue, catarrhal symptoms, and a fever (38.6 °C). Initial laboratory findings revealed hypoxemia (O2 saturation 79.8%), acidosis (pH 7.3), an elevated C-reactive protein (CRP) level of 14.8 mg/L, and a high D-dimer level (2.15 µg/mL). Nasopharyngeal (NP) antigen and RT-PCR tests confirmed SARS-CoV-2 infection, and an NP swab also detected penicillin- and ampicillin-resistant Staphylococcus aureus. She was admitted for conservative management, including oxygen supplementation, IV fluids, and prophylactic anticoagulation. Subsequently, she developed neurological symptoms—lethargy, discoordination, and impaired communication—without signs of meningism. Cerebrospinal fluid (CSF) analysis identified SARS-CoV-2 RNA (Ct = 29) on RT-PCR, while bacterial cultures remained negative. Treatment was intensified to include 10% mannitol, dexamethasone, and empiric ceftriaxone. Despite these interventions, the patient remained somnolent, with a Glasgow Coma Scale (GCS) score of 10. Upon discharge, her GCS had improved to 14; however, she continued to experience lethargy and cognitive issues, commonly described as “brain fog”. Inflammatory markers remained elevated (CRP 23 mg/L) and repeat RT-PCR of CSF confirmed a persistent SARS-CoV-2 presence (Ct = 31). This case underscores the potential for SARS-CoV-2 to cause prolonged CNS involvement, leading to persistent neurological impairment despite standard therapy. Further research is essential to clarify the pathophysiology of and determine optimal management for SARS-CoV-2 neuroinfections. Full article
(This article belongs to the Special Issue COVID-19 Complications and Co-infections)
Show Figures

Figure 1

Figure 1
<p>Cycle threshold (Ct) values for target genes in nasopharyngeal (NP) swabs and cerebrospinal fluid (CSF) samples at initial presentation and discharge. (<b>A</b>) Initial NP Swab Ct Values: RdRp = 19.910, E gene = 19.168, and N gene = 20.105. (<b>B</b>) Initial CSF Ct Values: RdRp = 29.074, E gene = 28.121, and N gene = 28.449. (<b>C</b>) NP Swab Ct Values at Discharge: E gene = 34.500 and N gene = 31.246. (<b>D</b>) CSF Ct Values at Discharge: RdRp = 31.900, E gene = 31.000, and N gene = 30.949.</p>
Full article ">Figure 2
<p>Chest X-ray at initial presentation.</p>
Full article ">
16 pages, 22416 KiB  
Article
A Combinatory Therapy of Metformin and Dexamethasone Reduces the Foreign Body Reaction to Intraneural Electrodes
by Bruno Rodríguez-Meana, Jaume del Valle and Xavier Navarro
Cells 2024, 13(24), 2112; https://doi.org/10.3390/cells13242112 - 20 Dec 2024
Viewed by 334
Abstract
Neural electrodes used for bidirectional communication between the nervous system and external devices like prosthetic limbs have advanced in neuroprosthetic applications. However, their effectiveness is hindered by the foreign body reaction, a natural immune response causing inflammation and fibrosis around the implanted device. [...] Read more.
Neural electrodes used for bidirectional communication between the nervous system and external devices like prosthetic limbs have advanced in neuroprosthetic applications. However, their effectiveness is hindered by the foreign body reaction, a natural immune response causing inflammation and fibrosis around the implanted device. This process involves protein adsorption, immune cell recruitment, cytokine release, and fibroblast activation, leading to a fibrous capsule formation and a decrease in electrode functionality. Anti-inflammatory and antifibrotic strategies have the potential to diminish the impact of the foreign body response. In this work, we have evaluated long-term metformin administration and short-term dexamethasone administration as a combined therapy to modulate the foreign body reaction induced by a polyimide intraneural implant in the sciatic nerve of rats. After a 12-week implant, the foreign body reaction was significantly reduced only in the group administered both drugs. Full article
(This article belongs to the Section Cells of the Nervous System)
Show Figures

Figure 1

Figure 1
<p>Results of the functional tests in rats with a PI device implanted in the tibial nerve. (<b>A</b>) Algesimetry test results expressed as percentages of force thresholds for withdrawal (vs. contralateral control paw) of animals before the implantation and after the implantation and treatments for 12 weeks. (<b>B</b>) The plot of the SFI obtained in the walking track test. No significant differences were found.</p>
Full article ">Figure 2
<p>Results of the functional tests in rats with a PI intraneural device implanted in the tibial nerve. Motor nerve conduction parameters of animals before implantation (Pre) and after the implantation of PI devices for 12 weeks and drug administration. (<b>A</b>,<b>B</b>) CMAP amplitudes of GM (<b>A</b>) and PL (<b>B</b>) muscles. (<b>C</b>,<b>D</b>) CMAP onset latencies of GM (<b>C</b>) and PL (<b>D</b>) muscles. No significant differences were found in electrophysiological test results.</p>
Full article ">Figure 3
<p>The effect of drug administration on the FBR to intraneural implants. (<b>A</b>) The number of inflammatory Iba1+ cells in the tibial nerve of animals implanted with PI devices and administered metformin, dexamethasone, or both. (<b>B</b>,<b>C</b>) Tissue capsule thickness around the devices in the tibial nerve of animals implanted with PI receiving the different treatments. Measurements were made using immunofluorescence sections (<b>B</b>) and thin sections of epon-embedded nerves (<b>C</b>). (<b>D</b>–<b>F</b>) Correlation between the number of Iba1+ cells and capsule thickness (IF) at 2, 8, and 12 weeks after implantation. The solid lines represent the linear regression, while the shaded area represents the 95% CIL. * <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, and ### <span class="html-italic">p</span> &lt; 0.01 time variable, two-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 4
<p>Representative images of inflammatory cells (red, Iba 1+ cell) infiltrating the tibial nerve after 2, 8, and 12 weeks of the PI intraneural device implantation in the different groups studied. Note the intense fluorescence emitted by the PI. The area limited by the dotted line corresponds to the tibial fascicle of the sciatic nerve that was used to analyze the number of labeled cells. Scale bar: 100 μm.</p>
Full article ">Figure 5
<p>Representative images of nerve cross-sections around the PI intraneural implant after 2, 8, and 12 weeks of the implantation in the different groups studied. Nerve fibers are labeled with antibody RT97. Note the intense fluorescence emitted by the PI. The measured capsule surrounding the PI device is the area delimited by the dotted line, which separates the PI from the nerve fibers, excluding tissue-empty regions. Scale bar: 50 μm.</p>
Full article ">Figure 6
<p>Representative images of cross-sections of the nerves embedded in epon resin and stained with toluidine blue, corresponding to samples taken at 2, 8, and 12 weeks for the different study groups. The images show the PI implants (pointed to by a red arrow in the top-right panel) within the nerve, surrounded by the capsule and axons. The thickness of the capsule from the implant to the first axons is marked with a red bar in the top-right panel. Images were acquired and transformed to greyscale. Scale bar: 50 μm for all the panels.</p>
Full article ">Figure 7
<p>Representative images of the capsule composition around the PI intraneural implant. Immunohistochemical labeling for macrophages (red, Iba 1+), fibroblasts (green, CD90, arrowheads), and nuclei (blue, DAPI) of tibial nerves of animals of the different groups implanted with a PI device after 2, 8, and 12 weeks. Scale bar: 10 μm. Images with the individual channels are presented as <a href="#app1-cells-13-02112" class="html-app">Supplementary Materials Figures S1–S3</a>.</p>
Full article ">Figure 8
<p>Representative images of nerve sections stained with Masson’s trichrome stain, showing the deposition of collagen in the capsule around the PI intraneural implant. At 2 weeks, the pink-stained area, outlined by the dotted line, corresponds to macrophages around the implant. At 8 and 12 weeks, the pink areas around the devices decreased, while the blue-stained areas (dotted line), composed of collagen fibers, were more preeminent surrounding the implant. Scale bar: 50 and 20 μm.</p>
Full article ">
14 pages, 3804 KiB  
Article
Pre-Adipocytes in 3D Co-Culture Underwent Self-Differentiation: New Perspectives for an Old Model
by Tamara Dal-Mora, Najla Adel Saleh, Veridiana Pacheco Goulart Martinazzo, Maria Luiza Carneiro Buchele, Michele Patrícia Rode, Adny Henrique Silva, Laura Sartori Assunção, Tânia Beatriz Creczynski-Pasa and Fabiola Branco Filippin-Monteiro
Organoids 2024, 3(4), 295-308; https://doi.org/10.3390/organoids3040018 - 18 Dec 2024
Viewed by 400
Abstract
Adipogenesis is a complex process influenced by various cellular interactions within adipose tissue, which plays a critical role in metabolic homeostasis. This study aimed to develop a novel in vitro three-dimensional (3D) co-culture model using murine 3T3-L1 pre-adipocytes, J774 macrophages, and NIH-3T3 fibroblasts [...] Read more.
Adipogenesis is a complex process influenced by various cellular interactions within adipose tissue, which plays a critical role in metabolic homeostasis. This study aimed to develop a novel in vitro three-dimensional (3D) co-culture model using murine 3T3-L1 pre-adipocytes, J774 macrophages, and NIH-3T3 fibroblasts to investigate adipogenic differentiation and inflammatory pathways. We first validated an adipogenic differentiation protocol in a two-dimensional (2D) model, where 3T3-L1 pre-adipocytes were subjected to a hormonal medium containing 3-isobutyl-1-methylxanthine, dexamethasone and insulin. After 7 days, differentiated cells were analyzed using Oil Red O and Nile Red staining, confirming lipid accumulation. Subsequently, spheroids were formed in 3D cultures, with monospheroids and heterospheroids maintained in either control medium or MDI for 11 days. Size measurements indicated significant growth in heterospheroids, particularly in the 3T3-L1:J774 combination, underscoring the importance of cellular interactions. Confocal microscopy and flow cytometry analyses demonstrated that even in the absence of hormonal stimuli, control spheroids exhibited adipogenic differentiation, evidenced by a notable proportion of Nile Red-positive cells (75.7 ± 1.7%). Inflammatory profiling revealed that the heterospheroid 3:J produced the highest levels of nitric oxide (NO), with no significant differences observed between control and MDI conditions. This study highlights the potential of 3D co-culture systems for elucidating the intricate interactions among adipocytes, macrophages, and fibroblasts. The findings may provide valuable insights into novel therapeutic targets for metabolic disorders. Full article
Show Figures

Figure 1

Figure 1
<p>Spheroid growth and cell viability. The model for the spheroid formation process (<b>A</b>). Representative images of spheroids formation in control (<b>B</b>) and MDI medium (<b>C</b>). Scale bar: 100 µm. Data are expressed as the mean ± standard deviation (SD) of three experiments. One-way ANOVA, followed by Dunnett’s multiple comparisons test, was used for statistical significance. Growth of spheroids formed on control medium (<b>D</b>). *** (<span class="html-italic">p</span> &lt; 0.001) 3:J:N group in relation to all other groups on day 4. ɛɛɛ (<span class="html-italic">p</span> &lt; 0.001) 3:J:N group in relation to all other groups except 3:J on day 7. ɸɸɸ (<span class="html-italic">p</span> &lt; 0.001) 3:J:N group in relation to all other groups except 3:J on day 11. Growth of spheroids formed on MDI medium (<b>E</b>). *** (<span class="html-italic">p</span> &lt; 0.001) 3:J:N group in relation to all other groups on day 4. ɛɛɛ (<span class="html-italic">p</span> &lt; 0.001) 3:J:N group in relation to N group on day 7. ɸɸɸ (<span class="html-italic">p</span> &lt; 0.001) 3:J:N group in relation to N group on day 11. δ (<span class="html-italic">p</span> &lt; 0.05) 3:J:N group in relation to 3:J group on day 11. Legend: 3 = 3T3-L1; N = NIH-3T3; 3:J = 3T3-L1:J774; 3:N = 3T3-L1:NIH-3T3; 3:J:N = 3T3-L1:J774:NIH-3T3.</p>
Full article ">Figure 2
<p>External morphology and cellular viability of different spheroids formed on day 11. SEM-imagens of spheroids formed in control (<b>A</b>) and MDI medium (<b>B</b>). Scale bar 100 and 10 µm. Legend: 3 = 3T3-L1; N = NIH-3T3; 3:J = 3T3-L1:J774; 3:N = 3T3-L1:NIH-3T3; 3:J:N = 3T3-L1:J774:NIH-3T3.</p>
Full article ">Figure 3
<p>Adipogenic differentiation of mono and heterospheroids with or without hormonal stimuli. Maximum intensity projection from 1 µm apart of spheroids formed in control (<b>A</b>) and MDI medium (<b>B</b>). Spheroids were stained with Nile red (yellow), and nuclei were counterstained with DAPI (blue). Scale bar 100 µm. Graphic representation of the percentage of positive Nile red cells (<b>C</b>). Data are expressed as mean ± SD of three experiments. Statistical analyses by Student’s <span class="html-italic">t</span>-test. Legend: 3 = 3T3-L1; N = NIH-3T3; 3:J = 3T3-L1:J774; 3:N = 3T3-L1:NIH-3T3; 3:J:N = 3T3-L1:J774:NIH-3T3.</p>
Full article ">Figure 4
<p>˙NO release profile in the supernatant of spheroids maintained in control medium or treated with MDI. Data are presented as mean ± standard deviation (n = 3). Statistical analysis was performed using one-way ANOVA followed by Bonferroni post hoc test. * (<span class="html-italic">p</span> &lt; 0.05) compared to the 3:J model; α (<span class="html-italic">p</span> &lt; 0.05) compared to the 3 model; β (<span class="html-italic">p</span> &lt; 0.05) compared to the N model; ϕ (<span class="html-italic">p</span> &lt; 0.05), ϕϕ (<span class="html-italic">p</span> &lt; 0.01) compared to the 3:J. 3 = 3T3-L1; N = NIH/3T3; 3:J = 3T3-L1; 3:N = 3T3-L1/3T3; 3:J:N = 3T3-L1:J774/3T3 model.</p>
Full article ">
13 pages, 1507 KiB  
Article
Potential Effect of Defatted Mealworm Hydrolysate on Muscle Protein Synthesis in C2C12 Cells and Rats
by Seo-Hyun Choi, Tae-Hwan Jung and Kyoung-Sik Han
Appl. Sci. 2024, 14(24), 11772; https://doi.org/10.3390/app142411772 - 17 Dec 2024
Viewed by 340
Abstract
(1) Background: the objective of this study was to examine the impact of defatted mealworm hydrolysate (DMH), formulated through protein hydrolysis, on muscle protein synthesis in C2C12 cells and rats; (2) Methods: C2C12 cells were treated with dexamethasone and DMH, and cell viability [...] Read more.
(1) Background: the objective of this study was to examine the impact of defatted mealworm hydrolysate (DMH), formulated through protein hydrolysis, on muscle protein synthesis in C2C12 cells and rats; (2) Methods: C2C12 cells were treated with dexamethasone and DMH, and cell viability was quantified using the MTT assay. Twenty-four Sprague–Dawley rats were divided into three groups (control, DEX, and DEX + DMH) and treated for 8 weeks. The DEX and DEX + DMH groups were administered intraperitoneal injections of DEX at a concentration of 2.25 mg/kg over a 3-d period. The control and DEX groups were fed a control diet, whereas the DMH group had part of the protein composition of the control diet replaced with 3.5% of DMH. The impact of DMH on muscle protein synthesis was evaluated through the measurement of grip strength, gastrocnemius and tibialis anterior muscle weights, and the investigation of muscle protein synthesis and degradation factor mRNA expression utilising the real-time PCR method; (3) Results: in vitro experiments demonstrated that treatment with DMH at concentrations greater than 5 mg/mL markedly alleviated DEX-induced injury in C2C12 cells. In vivo experiments demonstrated that the mRNA expression levels of myogenin and myoblast determination proteins, which promote muscle protein synthesis, were significantly increased. Furthermore, rats fed DMH exhibited significantly enhanced grip strength and tibialis anterior weight; (4) Conclusions: these findings indicate that DMH may serve as a functional material capable of promoting muscle protein synthesis and that the utilization of proteolytic enzymes is advantageous for the effective utilization of mealworms. Full article
(This article belongs to the Special Issue Functional Foods: Bioactivity and Potential Health Effects)
Show Figures

Figure 1

Figure 1
<p>Sodium dodecyl sulfate-polyacrylamide gel electrophoresis profile and non-protein nitrogen analysis of DMP and DMH. (<b>A</b>) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis; (<b>B</b>) non-protein nitrogen. Values are means ± standard error. The data were analyzed using Student’s <span class="html-italic">t</span>-test (* <span class="html-italic">p</span> &lt; 0.05). M, molecular weight marker; DMP, defatted mealworm powder; DMH, defatted mealworm hydrolysate.</p>
Full article ">Figure 2
<p>Cell viability of DMH on dexamethasone-induced C2C12 cell viability. (<b>A</b>) Cytotoxicity of DMH; (<b>B</b>) cell protective effect of DMH. Values are means ± standard error. The data were analyzed using the Student’s <span class="html-italic">t</span>-test (* <span class="html-italic">p</span> &lt; 0.05) compared to the group without DMH added. DMH, defatted mealworm hydrolysate. DEX, dexamethasone.</p>
Full article ">Figure 3
<p>Body weight change of rats during the experiment. Values are means ± standard error. Different letters (a and b) above the bars indicate significant differences by one-way analysis of variance with Duncan’s multiple range test (<span class="html-italic">p</span> &lt; 0.05). DEX, dexamethasone (2.25 mg/kg, i.p.) + control diet; DEX + DMH, dexamethasone (2.25 mg/kg, i.p.) + defatted mealworm hydrolysate diet; i.p., intraperitoneal injection.</p>
Full article ">Figure 4
<p>Grip strength test of rats fed the control and experimental diets for 8 weeks. The data were analyzed using the Student’s <span class="html-italic">t</span>-test (* <span class="html-italic">p</span> &lt; 0.05) compared to control. DEX, dexamethasone (2.25 mg/kg, intraperitoneal injection) + control diet; DEX + DMH, dexamethasone (2.25 mg/kg, intraperitoneal injection) + defatted mealworm hydrolysate diet.</p>
Full article ">Figure 5
<p>Weight of muscle tissue in rats. Values are means ± standard error. The data were analyzed using the Student’s <span class="html-italic">t</span>-test (* <span class="html-italic">p</span> &lt; 0.05) compared to control. TA, tibialis anterior muscle; GAS, gastrocnemius muscle. DEX, dexamethasone (2.25 mg/kg, intraperitoneal injection) + control diet; DEX + DMH, dexamethasone (2.25 mg/kg, intraperitoneal injection) + defatted mealworm hydrolysate diet.</p>
Full article ">Figure 6
<p>Expression of mRNA for muscle protein synthesis and degradation factors in the muscle tissue obtained from the rats. Values are means ± standard error. The data were analyzed using the Student’s <span class="html-italic">t</span>-test (* <span class="html-italic">p</span> &lt; 0.05) compared to DEX group. DEX, dexamethasone (2.25 mg/kg, intraperitoneal injection) + control diet; DEX + DMH, dexamethasone (2.25 mg/kg, intraperitoneal injection) + defatted mealworm hydrolysate diet; MuRF-1, muscle RING finger-1; MyoD, myoblast determination protein.</p>
Full article ">
18 pages, 31117 KiB  
Article
Synergistic Effects of Photobiomodulation and Differentiation Inducers on Osteogenic Differentiation of Adipose-Derived Stem Cells in Three-Dimensional Culture
by Daniella Da Silva, Anine Crous and Heidi Abrahamse
Int. J. Mol. Sci. 2024, 25(24), 13350; https://doi.org/10.3390/ijms252413350 (registering DOI) - 12 Dec 2024
Viewed by 409
Abstract
Osteoporosis, a common metabolic bone disorder, leads to increased fracture risk and significant morbidity, particularly in postmenopausal women and the elderly. Traditional treatments often fail to fully restore bone health and may cause side effects, prompting the exploration of regenerative therapies. Adipose-derived stem [...] Read more.
Osteoporosis, a common metabolic bone disorder, leads to increased fracture risk and significant morbidity, particularly in postmenopausal women and the elderly. Traditional treatments often fail to fully restore bone health and may cause side effects, prompting the exploration of regenerative therapies. Adipose-derived stem cells (ADSCs) offer potential for osteoporosis treatment, but their natural inclination toward adipogenic rather than osteogenic differentiation poses a challenge. This study investigates a novel approach combining differentiation inducers (DIs), three-dimensional (3D) hydrogel scaffolds, and photobiomodulation (PBM) to promote osteogenic differentiation of immortalised ADSCs. A dextran-based 3D hydrogel matrix, supplemented with a DI cocktail of dexamethasone, β-glycerophosphate disodium, and ascorbic acid, was used to foster osteogenesis. PBM was applied using near-infrared (825 nm), green (525 nm), and combined wavelengths at fluences of 3 J/cm2, 5 J/cm2, and 7 J/cm2 to enhance osteogenic potential. Flow cytometry identified osteoblast-specific markers, while inverted light microscopy evaluated cellular morphology. Reactive oxygen species assays measured oxidative stress, and quantitative polymerase chain reaction (qPCR) revealed upregulated gene expression linked to osteogenesis. The findings demonstrate that integrating DIs, 3D hydrogels, and PBM effectively drives osteogenic differentiation in immortalised ADSCs. The PBM enhanced osteogenic marker expression, induced morphological changes, and upregulated gene activity, presenting a promising framework for bone regeneration. Future research should assess the stability and functionality of these differentiated cells and explore their applicability in preclinical models of bone injury or degeneration. This integrative approach demonstrated specific efficacy in promoting the osteogenic differentiation of ADSCs, highlighting its potential application in developing targeted treatments for osteoporosis. Full article
(This article belongs to the Special Issue Regenerative Medicine: Biomaterials and Stem Cell Research)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Flow cytometric analysis of CD90 marker expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, a statistically significant increase in CD90 expression was observed in the G 5 J/cm<sup>2</sup> group (<span class="html-italic">p</span> &lt; 0.0001). (<b>b</b>) At 7 days post-treatment, CD90 expression declined in the G 5 J/cm<sup>2</sup> group, while a statistically significant increase was noted in the NIR 7 J/cm<sup>2</sup> group (<span class="html-italic">p</span> &lt; 0.0001) compared to the control. The data are expressed as mean ± SE. * <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; 0.0001. Black stars (*) indicate comparisons between the specified samples and the standard group. Blue stars (<span style="color:#0432FF">*</span>) denote comparisons between the experimental samples and the control group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 2
<p>Flow cytometric analysis of RUNX2 expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, RUNX2 expression showed a statistically significant increase in the NIR wavelength group at 3 J/cm<sup>2</sup> and 7 J/cm<sup>2</sup> fluencies (<span class="html-italic">p</span> &lt; 0.05) compared to the NIR 5 J/cm<sup>2</sup>, G 3 J/cm<sup>2</sup>, and G 7 J/cm<sup>2</sup> groups. (<b>b</b>) At 7 days post-treatment, RUNX2 expression significantly increased in the NIR 7 J/cm<sup>2</sup> group compared to the NIR 3 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.0001), NIR 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05), and G 7 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05) groups. The data are expressed as mean ± SE. * <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; 0.0001. Black stars (*) indicate comparisons between the specified samples and the standard group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 3
<p>Flow cytometric analysis of BGLAP expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, BGLAP expression was significantly increased in the NIR 5 J/cm<sup>2</sup> group compared to the NIR 3 J/cm<sup>2</sup> and NIR 7 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.01). (<b>b</b>) At 7 days post-treatment, no statistically significant BGLAP expression was observed in any experimental group. The data are expressed as mean ± SE. ** <span class="html-italic">p</span> &lt; 0.01. Black stars (*) indicate comparisons between the specified samples and the standard group. The sample size was n = 3.</p>
Full article ">Figure 4
<p>Flow cytometric analysis of BGN expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, BGN expression significantly increased in the G 5 J/cm<sup>2</sup> group compared to the G 3 J/cm<sup>2</sup>, G 7 J/cm<sup>2</sup>, NIR 5 J/cm<sup>2</sup>, and NIR-G 5 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.0001). (<b>b</b>) At 7 days post-treatment, a significant increase in BGN expression was observed in the NIR 5 J/cm<sup>2</sup> group compared to the G 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05) and NIR-G 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.0001) groups. Additionally, BGN expression in the NIR-G 7 J/cm<sup>2</sup> group was significantly higher compared to the NIR 7 J/cm<sup>2</sup> and G 7 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.0001). The data are expressed as mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. Black stars (*) indicate comparisons between the specified samples and the standard group. Blue stars (<span style="color:#0432FF">*</span>) denote comparisons between the experimental samples and the control group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 5
<p>Flow cytometric analysis of SOST expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, SOST expression showed a statistically significant increase in the G 5 J/cm<sup>2</sup> and NIR-G 5 J/cm<sup>2</sup> groups compared to the NIR 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.001), G 3 J/cm<sup>2</sup>, G 7 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.001), and NIR-G 3 J/cm<sup>2</sup> and NIR-G 7 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.0001). (<b>b</b>) At 7 days post-treatment, SOST expression remained significantly elevated in the G 5 J/cm<sup>2</sup> group compared to the NIR-G 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05), G 3 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.0001), and G 7 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.0001). The data are expressed as mean ± SE. * <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. Black stars (*) indicate comparisons between the specified samples and the standard group. Blue stars (<span style="color:#0432FF">*</span>) denote comparisons between the experimental samples and the control group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 6
<p>Morphological characterisation of immortalised adipose-derived mesenchymal stem cell differentiation at 24 h and 7 days following photobiomodulation treatment, observed with inverted light microscopy at 10× magnification.</p>
Full article ">Figure 7
<p>Increased intracellular reactive oxygen species production in immortalised adipose-derived mesenchymal stem cells at 24 h and 7 days following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, all experimental groups exhibited statistically significant increases in reactive oxygen species levels compared to the control group (<span class="html-italic">p</span> &lt; 0.0001), with the NIR-G combined wavelength group at 7 J/cm<sup>2</sup> showing a notable increase (<span class="html-italic">p</span> &lt; 0.05) compared to the NIR group at 7 J/cm<sup>2</sup>. (<b>b</b>) At 7 days post-treatment, significant increases in reactive oxygen species levels were observed in the NIR group at 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05), the G group at 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.01) and 7 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.001), and the NIR-G group at 7 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05), compared to the control group. The data are expressed as mean ± SE. * <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. Black stars (*) indicate comparisons between the specified samples and the standard group. Blue stars (<span style="color:#0432FF">*</span>) denote comparisons between the experimental samples and the control group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 8
<p>Quantitative PCR analysis of osteogenic gene expression. (<b>a</b>) At 24 h post-photobiomodulation treatment, CD90 was significantly downregulated (0.5-fold change) at a combined wavelength of 3 J/cm<sup>2</sup>, indicating a shift towards osteogenic differentiation. RUNX2 exhibited a pronounced upregulation, with a maximum fold change of 4.5 at 825 nm and 7 J/cm<sup>2</sup>, and additional increases of 2.3-fold at 525 nm with 3 J/cm<sup>2</sup> and 3.0-fold at 525 nm with 5 J/cm<sup>2</sup>. BGLAP was significantly upregulated (2.5-fold change) at 5 J/cm<sup>2</sup>, suggesting an early commitment to the osteogenic lineage. (<b>b</b>) At 7 days post-PBM treatment, CD90 showed notable upregulation at various PBM dosages, particularly at 825 nm with 3 J/cm<sup>2</sup> (2.4-fold change) and 7 J/cm<sup>2</sup> (4.4-fold change), as well as at 525 nm with 5 J/cm<sup>2</sup> (4.2-fold change). RUNX2 remained upregulated, with a 3.6-fold change at 825 nm and 3 J/cm<sup>2</sup> and a 3.9-fold change at 825 nm and 7 J/cm<sup>2</sup>. BGLAP continued to increase (2.6-fold change) at 825 nm and 7 J/cm<sup>2</sup>, while BGN showed significant upregulation (3.0-fold change) at 825 nm and 7 J/cm<sup>2</sup>. Additionally, SOST was upregulated (2.1-fold change) at 825 nm and 7 J/cm<sup>2</sup> and (2.8-fold change) at 525 nm and 5 J/cm<sup>2</sup>.</p>
Full article ">Figure 9
<p>Photobiomodulation treatment experimental design.</p>
Full article ">Figure 10
<p>Flow diagram of experimental design and methodology.</p>
Full article ">
12 pages, 2768 KiB  
Article
Prevention of Muscle Atrophy by Low-Molecular-Weight Fraction from Hirsutella sinensis Mycelium
by Yi-Wen Chen, Tsung-Ju Li, Li-Ching Wang, Bi-Hua Yang, Yen-Lien Chen, Chin-Chu Chen and Hsin-Tang Lin
Curr. Issues Mol. Biol. 2024, 46(12), 14033-14044; https://doi.org/10.3390/cimb46120839 - 12 Dec 2024
Viewed by 462
Abstract
Muscle atrophy, an age-related condition, presents a growing healthcare concern within the context of global population aging. While studies have investigated Hirsutella sinensis for its potential antifatigue properties, reports on its active components remain limited. This study evaluated the efficacy of H. sinensis [...] Read more.
Muscle atrophy, an age-related condition, presents a growing healthcare concern within the context of global population aging. While studies have investigated Hirsutella sinensis for its potential antifatigue properties, reports on its active components remain limited. This study evaluated the efficacy of H. sinensis mycelium extract on muscle health, utilizing a 1:1 water–ethanol preparation administered to C57BL/6 mice exhibiting acute hind leg atrophy. The results indicated no adverse effects, with significant improvements in muscle endurance and soleus muscle mass observed over a 14-day period. To further elucidate the mechanisms and effects of H. sinensis mycelium on dexamethasone-induced muscle atrophy, the water extract was fractionated into components of <3.5 kDa, 3.5–10 kDa, and >10 kDa using dialysis membranes. The investigation utilized a C2C12 cell atrophy model, induced by dexamethasone, to analyze the expression of relevant genes via qPCR. The results demonstrated that the <3.5 kDa and >10 kDa fractions significantly upregulated the expression of Myh2 and Myh7 genes while simultaneously downregulating the expression of MuRF-1 and Atrogin-1. It is noteworthy that the <3.5 kDa fraction exclusively enhanced MYHC protein expression and suppressed AMPK expression, as confirmed by Western blot analysis. This comprehensive pilot study suggests that the low-molecular-weight fraction of H. sinensis mycelium exhibits considerable potential for muscle mass preservation and atrophy mitigation. As a result, it offers a promising direction for the development of supplements aimed at addressing fatigue and preventing muscle atrophy. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of <span class="html-italic">H. sinensis</span> extract powder on IM-induced sarcopenia mice (<span class="html-italic">n</span> = 4). (<b>A</b>) Body weight; (<b>B</b>) food intake. N.S., no statistical significance.</p>
Full article ">Figure 2
<p>Effects of <span class="html-italic">H. sinensis</span> powder administration on grip strength and muscle endurance in mice (<span class="html-italic">n</span> = 4). (<b>A</b>) Grip strength after 7 days. (<b>B</b>) Grip strength after 14 days. (<b>C</b>) Muscle endurance after 7 days. (<b>D</b>) Muscle endurance after 14 days. Values with * have a significant difference (<span class="html-italic">p</span> &lt; 0.05); *** means <span class="html-italic">p</span> value is &lt;0.001; # means <span class="html-italic">p</span> value &lt; 0.05 compared to sham; N.S., no statistical significance.</p>
Full article ">Figure 3
<p>Effect of <span class="html-italic">H. sinensis</span> powder feeding for 14 days on the muscle mass of mice (<span class="html-italic">n</span> = 4) in the gastrocnemius and soleus muscles. (<b>A</b>) Gastrocnemius muscle. (<b>B</b>) Soleus muscle. Values with * have a significant difference (<span class="html-italic">p</span> &lt; 0.05) compared to vehicle; # means there is a significant difference (<span class="html-italic">p</span> &lt; 0.05) compared to sham.</p>
Full article ">Figure 4
<p>Evaluation of the efficacy of different solvent extracts of <span class="html-italic">H. sinensis</span>.</p>
Full article ">Figure 5
<p>Effects of different-molecular-weight fractions on C2C12 cell viability: (<b>A</b>) &lt;3.5 kDa, (<b>B</b>) 3.5–10 kDa, and (<b>C</b>) &gt;10 kDa. Results are expressed as mean ± SD (<span class="html-italic">n</span> = 3). N.S., no statistical significance.</p>
Full article ">Figure 6
<p>Gene expression levels in sarcopenic cells treated with different-molecular-weight fractions of <span class="html-italic">H. sinensis</span> aqueous extract. (<b>A</b>) <span class="html-italic">Myh2;</span> (<b>B</b>) <span class="html-italic">Myh7;</span> (<b>C</b>) <span class="html-italic">MuRF-1;</span> (<b>D</b>) <span class="html-italic">Atrogin-1</span>. Results are expressed as mean ± SD (<span class="html-italic">n</span> = 3). Compared with the dexamethasone group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001. Compared with the control group, # <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 7
<p>Effects of different-molecular-weight fractions of <span class="html-italic">H. sinensis</span> aqueous extract on MYHC and AMPK protein expression in sarcopenic C2C12 cells. (<b>A</b>) The results of protein expression in different extract fractions; (<b>B</b>) MYHC; (<b>C</b>) AMPK. Results are expressed as mean ± SD (<span class="html-italic">n</span> = 3). Compared with the control group, # <span class="html-italic">p</span> &lt; 0.05. Compared with the dexamethasone group, * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
18 pages, 4101 KiB  
Article
Maternal Low-Protein Diet During Nursing Leads to Glucose–Insulin Dyshomeostasis and Pancreatic-Islet Dysfunction by Disrupting Glucocorticoid Responsiveness in Male Rats
by Paulo Cezar de Freitas Mathias, Aline Milena Dantas Rodrigues, Patrícia Cristina Lisboa, Rosiane Aparecida Miranda, Ananda Malta, Tatiane Aparecida Ribeiro, Luiz Felipe Barella, Ginislene Dias, Thalyne Aparecida Leite Lima, Rodrigo Mello Gomes, Egberto Gaspar de Moura and Júlio Cezar de Oliveira
Biology 2024, 13(12), 1036; https://doi.org/10.3390/biology13121036 - 11 Dec 2024
Viewed by 518
Abstract
Both perinatal malnutrition and elevated glucocorticoids are pivotal triggers of the growing global pandemic of metabolic diseases. Here, we studied the effects of metabolic stress responsiveness on glucose–insulin homeostasis and pancreatic-islet function in male Wistar offspring whose mothers underwent protein restriction during lactation. [...] Read more.
Both perinatal malnutrition and elevated glucocorticoids are pivotal triggers of the growing global pandemic of metabolic diseases. Here, we studied the effects of metabolic stress responsiveness on glucose–insulin homeostasis and pancreatic-islet function in male Wistar offspring whose mothers underwent protein restriction during lactation. During the first two weeks after delivery, lactating dams were fed a low-protein (4% protein, LP group) or normal-protein diet (22.5% protein, NP group). At 90 days of age, male rat offspring were challenged with food deprivation (72 h of fasting), intracerebroventricular (icv) injection of dexamethasone (2 µL, 2.115 mmol/L) or chronic intraperitoneal injection of dexamethasone (1 mg/kg body weight/5 days). Body weight, food intake, intravenous glucose tolerance test (ivGTT) results, insulin secretion and biochemical parameters were assessed. LP rats did not display significant metabolic changes after long-term starvation (p > 0.05) or under the central effect of dexamethasone (p = 0.999). Chronic dexamethasone induced rapid hyperglycemia (~1.2-fold, p < 0.001) and hyperinsulinemia (NP: 65%; LP: 216%; p < 0.001), decreased insulin sensitivity (NP: ~2-fold; LP: ~4-fold; p < 0.001), reduced insulinemia (20%) and increased glycemia (35%) only in NP rats under ivGTT conditions (p < 0.001). Glucose and acetylcholine insulinotropic effects, as well as the muscarinic receptor antagonist response, were reduced by chronic dexamethasone only in pancreatic islets from NP rats (p < 0.05). The direct effect of dexamethasone on pancreatic islets reduced insulin secretion (NP: 60.2%, p < 0.001; LP: 33.8%, p < 0.001). Peripheral glucose–insulin dyshomeostasis and functional failure of pancreatic islets in LP rats, as evidenced by an impaired acute and chronic response to metabolic stress, may be due to excessive corticosterone action as a long-term consequence. Full article
(This article belongs to the Special Issue β-Cells at the Center of Type 1 and Type 2 Diabetes)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Body weight and food intake assessment throughout the chronic dexamethasone treatment. The data are presented as the means ± SEMs of 12 rats from 8 litters. Body weight evolution of NP (<b>A</b>) and LP (<b>B</b>) rats and food intake of NP (<b>D</b>) and LP (<b>E</b>) rats during dexamethasone treatment. The significant differences among the points in the curves (<b>A</b>,<b>B</b>,<b>D</b>,<b>E</b>) were analyzed via Student’s <span class="html-italic">t</span> test, and the values representing the area under the curve (AUC) for body weight (<b>C</b>) and food intake (<b>F</b>) were analyzed via one-way ANOVA, followed by Tukey’s post hoc test. * <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 denote significant differences versus NP, and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 denote significant differences versus LP. NP, normal-protein diet; LP, low-protein diet.</p>
Full article ">Figure 2
<p>Ad libitum food intake during the dark cycle after icv injection of dexamethasone. The data are presented as the means ± SEMs of 12 h fasted (7:00 a.m.–7:00 p.m.) 8 rats obtained from 8 litters. The values were assessed 4 h (at 11:00 p.m.) and 12 h (at 7:00 a.m.) after saline or dexamethasone intracerebroventricular (icv) injection. The significant differences were analyzed with one-way ANOVA, followed by Tukey’s post hoc test, for 4 h and 12 h. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 denotes significant differences versus NP, and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 denotes a significant difference versus LP. NP, normal-protein diet; LP, low-protein diet.</p>
Full article ">Figure 3
<p>Variation in plasma insulin levels during the intravenous glucose tolerance test (ivGTT). The data are presented as the means ± SEMs of 8 rats from 8 litters. Plasma insulin levels of NP and LP rats without stress (<b>A</b>,<b>B</b>) and of NP (<b>C</b>,<b>D</b>) and LP (<b>E</b>,<b>F</b>) rats after 72 h of starvation and/or chronic dexamethasone challenge. The significant differences were analyzed with Student’s <span class="html-italic">t</span> test (<b>A</b>,<b>B</b>) or one-way ANOVA, followed by Tukey’s post hoc test (<b>C</b>–<b>F</b>). * <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 denote significant differences versus NP/Sal rats, and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 denotes significant differences versus LP/Sal rats. NP, normal-protein diet; LP, low-protein diet.</p>
Full article ">Figure 4
<p>Variation in plasma glucose levels during the intravenous glucose tolerance test (ivGTT). The data are presented as the means ± SEMs of 8 rats from 8 litters. Plasma glucose levels of NP and LP rats without stress (<b>A</b>,<b>B</b>) and of NP (<b>C</b>,<b>D</b>) and LP (<b>E</b>,<b>F</b>) rats after 72 h of starvation and/or chronic dexamethasone challenge. The significant differences were analyzed with Student’s <span class="html-italic">t</span> test (<b>A</b>,<b>B</b>) or one-way ANOVA, followed by Tukey’s post hoc test (<b>C</b>–<b>F</b>). * <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 denote significant differences versus NP/Sal rats, and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 denote significant differences versus LP/Sal rats. NP, normal-protein diet; LP, low-protein diet.</p>
Full article ">Figure 5
<p>Insulin secretion from isolated pancreatic islets under the insulinotropic action of different glucose (5.6, 8.3, 11.1, 16.7, 20.0 and 24.0 mmol/L: (<b>A</b>–<b>C</b>)) and acetylcholine (0.1, 1.0, 10.0, 100.0 and 1000.0 µmol/L: (<b>D</b>,<b>F</b>)) concentrations. The symbols in the curves represent the means ± SEMs of insulin release from NP versus LP rats without stress challenges (<b>A</b>,<b>D</b>), from NP rats without stress challenges versus NP rats with chronic dexamethasone challenge (<b>B</b>,<b>E</b>) and from LP rats without stress challenges versus LP rats with chronic dexamethasone challenge (<b>C</b>,<b>F</b>). A pool of pancreatic islets (n = 32 islets) was obtained from 4 rats from 4 different litters in each experimental group. The significant differences between the groups for each glucose (<b>A</b>–<b>C</b>) and acetylcholine (<b>D</b>–<b>F</b>) concentration were determined by Student’s <span class="html-italic">t</span> test. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 denote significant differences versus the NP/Sal group, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 denote significant differences versus the LP/Sal group. NP/Sal, NP rats treated with saline; NP/Dex, NP rats treated with dexamethasone; LP/Sal, LP rats treated with saline; LP/Dex, LP rats treated with dexamethasone. NP, normal-protein diet; LP, low-protein diet.</p>
Full article ">Figure 6
<p>Insulin secretion from isolated pancreatic islets under the action of a selective antagonist of the insulinotropic subtype 3 muscarinic acetylcholine receptor (M<sub>3</sub>mAChR) (<b>A</b>) and different dexamethasone concentrations (1, 2, 4, 8 and 16 µmol/L) (<b>B</b>). The data are presented as the means ± SEMs of insulin release. A pool of pancreatic islets (n = 32 islets) was obtained from 4 rats from 4 different litters in each experimental group. The significant differences between the groups for each insulin secretagogue agent (glucose, 8.3 mmol/L; acetylcholine, 10 µmol/L; or 4-DAMP, 100 µmol/L) and dexamethasone in the presence of 16.7 mmol/L glucose were determined by one-way ANOVA, followed by Tukey’s post hoc test. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 denote significant differences between each of the groups (LP/Sal, NP/Dex and LP/Dex) versus the NP/Sal group with each of the treatments (Glu, ACh and 4-DAMP) and for all the dexamethasone concentrations versus 16.7 mmol/L glucose-induced insulin secretion in the NP/Sal islets. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 denotes a significant difference between the dexamethasone concentrations and 16.7 mmol/L glucose-induced insulin secretion in LP/Sal islets. NP/Sal, NP rats treated with saline; NP/Dex, NP rats treated with dexamethasone; LP/Sal, LP rats treated with saline; LP/Dex, LP rats treated with dexamethasone. NP, normal-protein diet; LP, low-protein diet. Glu, glucose; ACh, acetylcholine; 4-DAMP, 4-diphenylacetoxy-N-methylpiperidine methiodide; Dex, dexamethasone; + indicates the presence or absence ⃝ of insulin secretagogues (ACh and 4-DAMP) or dexamethasone.</p>
Full article ">
30 pages, 6814 KiB  
Article
Effects of Alnus japonica Hot Water Extract and Oregonin on Muscle Loss and Muscle Atrophy in C2C12 Murine Skeletal Muscle Cells
by Da Hyeon An, Chan Ho Lee, Yeeun Kwon, Tae Hee Kim, Eun Ji Kim, Jae In Jung, Sangil Min, Eun Ju Cheong, Sohyun Kim, Hee Kyu Kim and Sun Eun Choi
Pharmaceuticals 2024, 17(12), 1661; https://doi.org/10.3390/ph17121661 - 10 Dec 2024
Viewed by 527
Abstract
Background/Objectives: Sarcopenia is characterized by the loss of muscle mass and function, increases in mortality rate, and risk of comorbidities in the elderly. This study evaluated the effects of Alnus japonica hot water extract (AJHW) and its active compound, oregonin, on muscle atrophy [...] Read more.
Background/Objectives: Sarcopenia is characterized by the loss of muscle mass and function, increases in mortality rate, and risk of comorbidities in the elderly. This study evaluated the effects of Alnus japonica hot water extract (AJHW) and its active compound, oregonin, on muscle atrophy and apoptosis in vitro. Methods: AJHW underwent phytochemical analysis. C2C12 cells were subjected to H2O2 and dexamethasone to induce oxidative stress and muscle loss, after which AJHW and oregonin were administered to assess their impacts on cell viability, apoptosis, muscle protein synthesis stimulation, and muscle protein degradation inhibition. Cell viability was assessed via an MTT assay, and apoptosis was analyzed by measuring Bcl-2, Bax, cleaved caspase-3, and cleaved PARP through Western blotting. Western blotting and RT-PCR were utilized to analyze MyoD, Myogenin, Atrogin-1, and MuRF1 protein and gene expression in a muscle atrophy model, as well as the Akt/mTOR and FoxO3α pathways. Results: AJHW was confirmed to contain oregonin, an active compound. AJHW and oregonin significantly increased cell viability and reduced apoptosis by upregulating Bcl-2 and downregulating Bax, cleaved caspase-3, and cleaved PARP. They significantly enhanced muscle protein synthesis through the upregulation of MyoD and Myogenin, while diminishing muscle degradation by downregulating Atrogin-1 and MuRF1. The activation of the Akt/mTOR pathway and inhibition of the FoxO3α pathway were also observed. Conclusions: AJHW and oregonin effectively prevented muscle cell apoptosis, promoted muscle protein synthesis, and inhibited muscle protein degradation in vitro. These results suggest that AJHW and oregonin could serve as therapeutic agents to prevent and treat sarcopenia. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>TLC chromatograms of each sample compound and the references: (<b>A</b>) UV 254 nm, (<b>B</b>) 10% H<sub>2</sub>SO<sub>4</sub>, (<b>C</b>) <span class="html-italic">ρ</span>-anisaldehyde H<sub>2</sub>SO<sub>4</sub>, and (<b>D</b>) FeCl<sub>3</sub>. The eluent system employed was chloroform/methanol/water = 70:30:4 (<span class="html-italic">v</span>/<span class="html-italic">v</span>/<span class="html-italic">v</span>). ① Oregonin, ② <span class="html-italic">Alnus japonica</span> ethanol extract (AJE), and ③ <span class="html-italic">Alnus japonica</span> hot water extract (AJHW).</p>
Full article ">Figure 2
<p>Negative mode LC-MS/MS analysis of oregonin (1000 μg/mL). (<b>A</b>) Extracted ion chromatogram of oregonin and (<b>B</b>) product ion mass spectrum of oregonin.</p>
Full article ">Figure 3
<p>Calibration curve and equation of oregonin (100, 50, 25, 10, 5, and 1 μg/mL of oregonin).</p>
Full article ">Figure 4
<p>HPLC chromatogram of oregonin (100, 50, 25, 10, 5, and 1 μg/mL of oregonin).</p>
Full article ">Figure 5
<p>HPLC chromatogram of (<b>A</b>) <span class="html-italic">Alnus japonica</span> ethanol extract (AJE) (1000 μg/mL) and (<b>B</b>) <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) (1000 μg/mL).</p>
Full article ">Figure 6
<p>Cytotoxicity of <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) on C2C12 myoblasts. (<b>A</b>) Treatment concentrations: 0, 25, 50, 100, 200, 400, 600, 800, and 1000 μg/mL. (<b>B</b>) Treatment concentrations: 0, 2.5, 5, 10, 20, and 25 μg/mL. Cell viability was calculated as described in <a href="#sec4-pharmaceuticals-17-01661" class="html-sec">Section 4</a>. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 5). *** <span class="html-italic">p</span> &lt; 0.001 significantly different from that of the 0 μg/mL group.</p>
Full article ">Figure 7
<p>Cytotoxicity of oregonin (ORE) on C2C12 myoblasts. Cell viability was calculated as outlined in the <a href="#sec4-pharmaceuticals-17-01661" class="html-sec">Section 4</a>. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 4). *** <span class="html-italic">p</span> &lt; 0.001 indicating a significant difference from the 0 μg/mL group.</p>
Full article ">Figure 8
<p>Protective effect of (<b>A</b>) <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and (<b>B</b>) oregonin (ORE) on cell viability in H<sub>2</sub>O<sub>2</sub>-treated C2C12 myoblasts. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 4). *** <span class="html-italic">p</span> &lt; 0.001 significantly different from that of [H<sub>2</sub>O<sub>2</sub> (−)/AJHW (−)] and [H<sub>2</sub>O<sub>2</sub> (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 significantly different from that of the [H<sub>2</sub>O<sub>2</sub> (+)/AJHW (−)] and [H<sub>2</sub>O<sub>2</sub> (+)/ORE (−)] group.</p>
Full article ">Figure 9
<p>Protective effect of (<b>A</b>) <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and (<b>B</b>) oregonin (ORE) on cell viability in DEX-treated C2C12 myotubes. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 significantly different from that of [DEX (−)/AJHW (−)], [DEX (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 significantly different from that of [DEX (+)/AJHW (−)], [DEX (+)/ORE (−)] group.</p>
Full article ">Figure 10
<p>Protective effect of (<b>A</b>) <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and (<b>B</b>) oregonin (ORE) on apoptosis in H<sub>2</sub>O<sub>2</sub>-treated C2C12 myoblasts. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 4). *** <span class="html-italic">p</span> &lt; 0.001 significantly different from that of [H<sub>2</sub>O<sub>2</sub> (−)/AJHW (−)], [H<sub>2</sub>O<sub>2</sub> (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 significantly different from those of the [H<sub>2</sub>O<sub>2</sub> (+)/AJHW (−)] and [H<sub>2</sub>O<sub>2</sub> (+)/ORE (−)] groups.</p>
Full article ">Figure 11
<p>Anti-apoptotic effects of <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and oregonin (ORE) on H<sub>2</sub>O<sub>2</sub>-induced oxidative damage in C2C12 myoblasts. Western blotting was used to analyze the levels of (<b>A</b>–<b>D</b>) Bax, (<b>E</b>–<b>H</b>) Bcl-2, and β-actin, as described in <a href="#sec4-pharmaceuticals-17-01661" class="html-sec">Section 4</a>. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 significantly different from that of [H<sub>2</sub>O<sub>2</sub> (−)/AJHW (−)], [H<sub>2</sub>O<sub>2</sub> (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 significantly different from those of the [H<sub>2</sub>O<sub>2</sub> (+)/AJHW (-)] and [H<sub>2</sub>O<sub>2</sub> (+)/ORE (−)] groups.</p>
Full article ">Figure 12
<p>Anti-apoptotic effects of <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and oregonin (ORE) on H<sub>2</sub>O<sub>2</sub>-induced oxidative damage in C2C12. Western blotting was used to analyze the levels of (<b>A</b>–<b>D</b>) cleaved caspase-3, (<b>E</b>–<b>H</b>) cleaved PARP, and β-actin, as outlined in <a href="#sec4-pharmaceuticals-17-01661" class="html-sec">Section 4</a>. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 significantly different from [H<sub>2</sub>O<sub>2</sub> (−)/AJHW (−)], [H<sub>2</sub>O<sub>2</sub> (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 significantly different from [H<sub>2</sub>O<sub>2</sub> (+)/AJHW (−)] and [H<sub>2</sub>O<sub>2</sub> (+)/ORE (−)] groups.</p>
Full article ">Figure 13
<p>Effects of (<b>A</b>,<b>C</b>) <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and (<b>B</b>,<b>D</b>) oregonin (ORE) on dexamethasone-induced muscle atrophy in C2C12 myotubes. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 5). The scale bar represents 100 μm. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 significantly different from that of [DEX (−)/AJHW (−)], [DEX (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 significantly different from that of [DEX (+)/AJHW (−)], [DEX (+)/ORE (−)] group.</p>
Full article ">Figure 14
<p>Effect of <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and oregonin (ORE) on the muscle-degradation-related protein expression levels of (<b>A</b>–<b>D</b>) Atrogin-1, (<b>E</b>–<b>H</b>) MuRF1 in DEX-treated C2C12 myotubes. AJHW and ORE were added to DEX-treated C2C12 myotubes and cultured for 24 h. Protein expression levels were determined using Western blotting. The expression levels were normalized to that of β-actin and expressed relative to those in the CON group. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 4). ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 significantly different from [DEX (−)/AJHW (−)], [DEX (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 significantly different from the [DEX (+)/AJHW (−)], [DEX (+)/ORE (−)] group.</p>
Full article ">Figure 15
<p>Effect of <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and oregonin (ORE) on the muscle-synthesis-related protein expression levels of (<b>A</b>–<b>D</b>) MyoD, and (<b>E</b>–<b>H</b>) Myogenin in DEX-treated C2C12 myotubes. AJHW and ORE were added to DEX-treated C2C12 myotubes and cultured for 24 h. Protein expression levels were determined using Western blotting. The protein expression levels were normalized to β-actin and expressed relative to those in the CON group. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 significantly different from that of [DEX (−)/AJHW (−)], [DEX (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 significantly different from the [DEX (+)/AJHW (−)], [DEX (+)/ORE (−)] group.</p>
Full article ">Figure 16
<p>Effect of <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and oregonin (ORE) on the protein expression levels of (<b>A</b>–<b>D</b>) phospho-Akt and Akt, and (<b>E</b>–<b>H</b>) phospho-mTOR and mTOR in DEX-treated C2C12 myotubes. AJHW and ORE were separately added to these cells and incubated for 24 h. Protein expression was assessed via Western blotting, with β-actin as the normalization standard, and expressed relative to the CON group. Values are reported as the mean ± S.E.M. (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, indicating significant differences from the [DEX (−)/AJHW (−)], [DEX (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 indicating significant differences from the [DEX (+)/AJHW (−)], [DEX (+)/ORE (−)] group.</p>
Full article ">Figure 16 Cont.
<p>Effect of <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and oregonin (ORE) on the protein expression levels of (<b>A</b>–<b>D</b>) phospho-Akt and Akt, and (<b>E</b>–<b>H</b>) phospho-mTOR and mTOR in DEX-treated C2C12 myotubes. AJHW and ORE were separately added to these cells and incubated for 24 h. Protein expression was assessed via Western blotting, with β-actin as the normalization standard, and expressed relative to the CON group. Values are reported as the mean ± S.E.M. (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, indicating significant differences from the [DEX (−)/AJHW (−)], [DEX (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 indicating significant differences from the [DEX (+)/AJHW (−)], [DEX (+)/ORE (−)] group.</p>
Full article ">Figure 17
<p>Effect of <span class="html-italic">Alnus japonica</span> hot water extract (AJHW) and oregonin (ORE) on the protein expression levels of (<b>A</b>–<b>D</b>) phospho-FoxO3α and FoxO3α in DEX-treated C2C12 myotubes. AJHW and ORE were separately added to these cells and incubated for 24 h. Protein expression was measured using Western blotting, normalized to β-actin, and reported relative to the CON group. Values are expressed as the mean ± S.E.M. (<span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 indicating significant differences from the [DEX (−)/AJHW (−)], [DEX (−)/ORE (−)] group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 indicating significant differences from the [DEX (+)/AJHW (−)], [DEX (+)/ORE (−)] group.</p>
Full article ">Figure 18
<p>The structure of oregonin. The chemical structure was illustrated using ChemDraw Ultra 7.0 (CambridgeSoft, Cambridge, MA, USA).</p>
Full article ">
15 pages, 3122 KiB  
Systematic Review
Clinical Efficacy of Cysteamine Application for Melasma: A Meta-Analysis
by Bing-Qi Wu, Yen-Jen Wang, Chang-Cheng Chang, Tzong-Yuan Juang, Yung-Hsueh Huang and Ying-Chuan Hsu
J. Clin. Med. 2024, 13(23), 7483; https://doi.org/10.3390/jcm13237483 - 9 Dec 2024
Viewed by 777
Abstract
Background: Melasma is a challenging, acquired hyperpigmentary disorder. The gold standard treatment is Kligman’s formulation, which contains hydroquinone, tretinoin, and dexamethasone, but its long-term use is limited by the risk of exogenous ochronosis. Cysteamine, a tyrosinase inhibitor, reduces melanocyte activity and melanin production, [...] Read more.
Background: Melasma is a challenging, acquired hyperpigmentary disorder. The gold standard treatment is Kligman’s formulation, which contains hydroquinone, tretinoin, and dexamethasone, but its long-term use is limited by the risk of exogenous ochronosis. Cysteamine, a tyrosinase inhibitor, reduces melanocyte activity and melanin production, showing strong depigmenting effects in patients resistant to Kligman’s formulation. Nonetheless, clinical studies have yielded inconsistent efficacy results. This meta-analysis aimed to assess the efficacy of cysteamine in treating melasma and to identify potential factors that may impact its therapeutic outcomes. Methods: A systematic search of PubMed, Embase, Web of Science, and CENTRAL, from the earliest record until August 2024, was conducted. Randomized controlled trials and quasi-randomized design studies related to topical cysteamine on melasma patients were included. The primary outcome was MASI or mMASI assessment after treatments. The current meta-analysis was conducted with a random-effects model. Subgroup analyses and meta-regressions were performed based on baseline MASI, disease duration of melasma, patient age, and sample size of the included studies. Funnel plots and Duval and Tweedie’s trim and fill method were adopted to assess the publication bias. Results: Eight studies were included for quantitative analysis. The analysis of MASI after topical cysteamine demonstrated a significant decrease compared to the placebo (p = 0.002). Compared to other melasma treatments, cysteamine did not show superior efficacy in mMASI (p = 0.277). The treatment efficacy of hydroquinone, modified Kligman’s formula, and tranexamic acid mesotherapy for melasma was not statistically different when compared to cysteamine (p = 0.434). Further analyses showed no benefit when allowing extended cysteamine application time (p < 0.0001). The meta-regression revealed the efficacy of cysteamine decreased as the duration of melasma increased (coefficient = 0.38, p = 0.0001, R2 = 0.99). The funnel plot displayed some asymmetry. The trim and fill method suggested the adjusted effect size was 0.607 (95% CI = −0.720 to 1.935). Conclusions: Cysteamine exhibited efficacy in treating melasma patients; however, its depigmentation effect was comparable to hydroquinone-based regimens, tranexamic acid mesotherapy, and modified Kligman’s formula. Using cysteamine in patients with a short duration of melasma may result in better efficacy. Full article
(This article belongs to the Special Issue Skin Diseases: From Diagnosis to Treatment)
Show Figures

Figure 1

Figure 1
<p>PRISMA 2020 flowchart for the current meta-analysis.</p>
Full article ">Figure 2
<p>Summary of quality assessment for the studies included in the current meta-analysis using version 2 of the Cochrane risk-of-bias tool for randomized trials [<a href="#B7-jcm-13-07483" class="html-bibr">7</a>,<a href="#B8-jcm-13-07483" class="html-bibr">8</a>,<a href="#B14-jcm-13-07483" class="html-bibr">14</a>,<a href="#B15-jcm-13-07483" class="html-bibr">15</a>,<a href="#B18-jcm-13-07483" class="html-bibr">18</a>,<a href="#B31-jcm-13-07483" class="html-bibr">31</a>,<a href="#B32-jcm-13-07483" class="html-bibr">32</a>,<a href="#B33-jcm-13-07483" class="html-bibr">33</a>].</p>
Full article ">Figure 3
<p>(<b>a</b>)<b>.</b> Forest plot presenting mean difference in depigmentation efficacy between cysteamine and placebo in patients with melasma. (<b>b</b>)<b>.</b> Forest plot presenting mean difference in depigmentation efficacy between cysteamine and other treatments in patients with melasma [<a href="#B7-jcm-13-07483" class="html-bibr">7</a>,<a href="#B8-jcm-13-07483" class="html-bibr">8</a>,<a href="#B14-jcm-13-07483" class="html-bibr">14</a>,<a href="#B15-jcm-13-07483" class="html-bibr">15</a>,<a href="#B18-jcm-13-07483" class="html-bibr">18</a>,<a href="#B31-jcm-13-07483" class="html-bibr">31</a>,<a href="#B32-jcm-13-07483" class="html-bibr">32</a>,<a href="#B33-jcm-13-07483" class="html-bibr">33</a>].</p>
Full article ">Figure 4
<p>In the efficacy of cysteamine compared to other treatments, a sensitivity analysis was performed using the one-study-removal method. The main result remained consistent without significant changes after excluding any of the included trials [<a href="#B7-jcm-13-07483" class="html-bibr">7</a>,<a href="#B15-jcm-13-07483" class="html-bibr">15</a>,<a href="#B18-jcm-13-07483" class="html-bibr">18</a>,<a href="#B31-jcm-13-07483" class="html-bibr">31</a>,<a href="#B32-jcm-13-07483" class="html-bibr">32</a>,<a href="#B33-jcm-13-07483" class="html-bibr">33</a>].</p>
Full article ">Figure 5
<p>The forest plot of subgroup analysis using different melasma treatments versus cysteamine as the moderator. The treatment efficacy of hydroquinone, modified Kligman’s formula, and tranexamic acid mesotherapy for melasma was not statistically different when compared to cysteamine. The Cochran’s Q test for the effect sizes difference among melasma treatment subgroups was insignificant (<span class="html-italic">p</span> = 0.434) [<a href="#B7-jcm-13-07483" class="html-bibr">7</a>,<a href="#B15-jcm-13-07483" class="html-bibr">15</a>,<a href="#B18-jcm-13-07483" class="html-bibr">18</a>,<a href="#B31-jcm-13-07483" class="html-bibr">31</a>,<a href="#B32-jcm-13-07483" class="html-bibr">32</a>,<a href="#B33-jcm-13-07483" class="html-bibr">33</a>].</p>
Full article ">Figure 6
<p>The forest plot of subgroup analysis is based on the permitted extended cysteamine application. In the study conducted by Lima et al., participants were asked to apply the cream for 15 min on the first night and gradually extend the duration up to 2 h. The Cochran’s Q test for the effect size differences among subgroups was significant (<span class="html-italic">p</span> &lt; 0.0001) [<a href="#B7-jcm-13-07483" class="html-bibr">7</a>,<a href="#B15-jcm-13-07483" class="html-bibr">15</a>,<a href="#B18-jcm-13-07483" class="html-bibr">18</a>,<a href="#B31-jcm-13-07483" class="html-bibr">31</a>,<a href="#B32-jcm-13-07483" class="html-bibr">32</a>,<a href="#B33-jcm-13-07483" class="html-bibr">33</a>].</p>
Full article ">Figure 7
<p>Meta-regression of difference in means on disease duration. The coefficient was 0.38 with <span class="html-italic">p</span> = 0.0001. Sachdev et al.’s study was omitted during the meta-regression due to the absence of exact disease duration in their trial.</p>
Full article ">Figure 8
<p>The funnel plot of included studies showed asymmetric distribution. The trim and fill method suggested the adjusted effect size was 0.607 (95% CI = −0.720 to 1.935).</p>
Full article ">
20 pages, 14462 KiB  
Article
Peripheral B Lymphocyte Serves as a Reservoir for the Persistently Covert Infection of Mandarin Fish Siniperca chuatsi Ranavirus
by Wenfeng Zhang, Hui Gong, Qianqian Sun, Yuting Fu, Xiaosi Wu, Hengwei Deng, Shaoping Weng, Jianguo He and Chuanfu Dong
Viruses 2024, 16(12), 1895; https://doi.org/10.3390/v16121895 - 9 Dec 2024
Viewed by 566
Abstract
Mandarin fish ranavirus (MRV) is a distinctive member among the genus Ranavirus of the family Iridoviridae. The persistently covert infection of MRV was previously observed in a natural outbreak of MRV, but the underlying mechanism remains unclear. Here, we show that mandarin [...] Read more.
Mandarin fish ranavirus (MRV) is a distinctive member among the genus Ranavirus of the family Iridoviridae. The persistently covert infection of MRV was previously observed in a natural outbreak of MRV, but the underlying mechanism remains unclear. Here, we show that mandarin fish peripheral B lymphocytes are implemented as viral reservoirs to maintain the persistent infection. When mandarin fish were infected with a sublethal dosage of MRV under a nonpermissive temperature (19 °C) and a permissive temperature (26 °C), all of the fish in the 19 °C group survived and entered the persistent phase of infection, characterized by a very low viral load in white blood cells, whereas some of the fish died of MRV infection in the 26 °C group, and the survival fish then initiated a persistent infection status. Raising the temperature, vaccination and dexamethasone treatment can reactivate the quiescent MRV to replicate and result in partial mortality. The viral reservoir investigation showed that IgM+-labeled B lymphocytes, but not CD3Δ+-labeled T lymphocytes and MRC-1+-labeled macrophages, are target cells for the persistent infection of MRV. Moreover, the reactivation of the quiescent MRV was confirmed through a non-TLR5 signal pathway manner. Collectively, we demonstrate the presence of the B cell-dependent persistent infection of ranavirus, and provide a new clue for better understanding the complex infection mechanism of vertebrate iridovirus. Full article
(This article belongs to the Section Animal Viruses)
Show Figures

Figure 1

Figure 1
<p>Tracking the status of MRV-infected mandarin fish under different temperature conditions. (<b>A</b>) The experimental outline of MRV infection and the quiescent model. Briefly, mandarin fish were infected under two different temperature conditions. In the 19 °C infection group, all of the fish survived to 30 dpi until raising the temperature stress. After acclimatization, the survival fish were treated with DXMS and heat-killed <span class="html-italic">E. coil</span>, respectively. In the 26 °C infection group, partial mortality occurred within 12 dpi. The survival fish were vaccinated with the inactivated MRV vaccine. (<b>B</b>) The survival curves of the MRV-infected mandarin fish under 19 °C and 26 °C. All of the fish survived in the 19 °C infection group, and partial mortality was observed in the 26 °C infection group. (<b>C</b>,<b>D</b>) The MRV copy number per mg WBC DNA isolated from the 20 mandarin fish in the 19 °C group and the 8 survival fish in the 26 °C group at 30 dpi, respectively. (<b>E</b>) Detection of MRV using the anti-MRV mAb-based colloidal gold-immunochromatographic fast-detection strip. A positive reaction was obtained only from the dead fish suffering from the primary infection of MRV in the 26 °C infection group (Acute), but negative for the 19 °C infection group and mock infection.</p>
Full article ">Figure 2
<p>Reactivation of the covert MRV via raising the temperature stress. (<b>A</b>) The experimental outline of the reactivation of covert MRV via raising the temperature stress. Blood was drawn from the infected fish in the 19 °C group for the MRV load analysis at 30 dpi. After 7 days of acclimatation, at 38 dpi, the water temperature was gradually elevated, with 1 °C per 2 days. During the raising of the temperature stress, dead fish were sampled for MRV load measurements, and the survival fish were used for the second round of blood collection for the MRV load assessment. (<b>B</b>) The MRV DNA load was measured in various tissues from 5 dead fish during the raising of the temperature stress. High MRV loads were observed in M2, M8 and M13. (<b>C</b>) The IHC of the intestines of the dead fish M13. The tissue was recognized with mAb 1C4, and numerous infected cells were stained brown by DAB. The dashed box indicates the enlarged area. Bar = 100 μm. (<b>D</b>,<b>E</b>) The MRV copy number per mg WBC DNA of each (<b>D</b>) and the gross distribution (<b>E</b>), respectively. Fifteen survival mandarin fish on the 5th day after the completion of the raising temperature stress and the comparison with that before stimulation. ***, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Assessment of the reactivation effects of covert MRV via vaccination, DXMS stimulation and <span class="html-italic">E. coli</span> injection. (<b>A</b>) Survival curve of the MRV-PCI mandarin fish via vaccination stimulation. (<b>B</b>) Tissue MRV DNA load examination of 3 dead fish suffering vaccination stimulation. (<b>C</b>) Survival curves of the MRV-PCI mandarin fish via DXMS treatment and <span class="html-italic">E. coli</span> injection. Three fish died during the DXMS treatment and no fish died during the <span class="html-italic">E. coil</span> injection. (<b>D</b>) Tissue MRV DNA load examination of 3 dead fish suffering from the DXMS treatment. (<b>E</b>–<b>G</b>) The repeated experiments (N = 20) of the assessment of the reactivation of MRV-PCI mandarin fish via vaccination and DXMS treatments, where 1, 7 and 10 fish died from the control, vaccination and DXMS treatments, respectively. High tissue MRV DNA loads were determined in dead fish from the vaccination (<b>F</b>) and DXMS treatment (<b>G</b>), respectively. Strong positive signals by IHC were also observed in the pyloric caeca from the vaccination (<b>H</b>) and DXMS treatment (<b>I</b>), respectively. The dashed box indicates the enlarged area. Bar = 100 μm.</p>
Full article ">Figure 4
<p>Detection of the antibody-targeted cell markers from mandarin fish WBCs. (<b>A</b>) Western blotting analysis of four cell markers of mandarin fish WBCs. The mAb of 7F12F6 recognized the light chain of the mandarin fish IgM. Three pAbs also recognized the corresponding protein bands, as speculated. (<b>B</b>,<b>C</b>) Confocal micrographs of mandarin fish WBCs. (<b>B1</b>) A few IgM<sup>+</sup>-labeled B cells from native WBCs. (<b>B2</b>) Numerous IgM<sup>+</sup>-labeled B cells after the flow cytometry sorting. (<b>B3</b>) Almost no IgM<sup>+</sup>-labeled B cells in the residual WBCS out of the B cells. Bar = 20 μm. (<b>C</b>) The enlarged image of the sorted IgM<sup>+</sup>-labeled B cells by the FACS. Bar =10 μm.</p>
Full article ">Figure 5
<p>Fluorescence-activated cell staining (FACS) sorting of mandarin fish peripheral WBCs. (<b>A</b>) Flow cytometry of the mandarin fish WBCs stained with antibodies to the mandarin fish IgM (left), CD3 (middle) and MRC1 (right). (<b>B</b>) Confocal micrographs of the mandarin fish IgM<sup>+</sup> and IgM<sup>−</sup> WBCs. IgM<sup>+</sup> cells were identified by anti-mandarin fish IgM mAb (7F12F6) and secondary Alexa Fluor555-labeled goat anti-mouse IgG (red). Pax5<sup>+</sup> cells were identified by anti-Pax5 pAb and secondary Alexa Fluor488-labeled goat anti-rabbit IgG (green). The nucleus was identified with DAPI (blue). Bar = 10 μm. (<b>C</b>,<b>D</b>) show the progress of the IgM<sup>+</sup> WBCs selected by the FACS and the copy number of the MRV genomes determined from 1 μg of DNA from the total WBCs, the selected IgM<sup>+</sup> B cells and the residual IgM<sup>−</sup> WBCs from the MRV-PCI mandarin fish, respectively. (<b>D</b>–<b>F</b>) The progress of the CD3<sup>+</sup> WBCs selected by the FACS, and the copy number of the MRV genomes detected from 1 μg of WBC DNA, selected CD3<sup>+</sup> T cells and CD3<sup>−</sup> WBCs from the MRV-PCI mandarin fish, respectively. (<b>G</b>,<b>H</b>) The MRC1<sup>+</sup> WBCs selected by the FACS (<b>G</b>) and the copy number of the MRV genomes from the WBCs, selected MRC1<sup>+</sup> Mø and MRC1<sup>−</sup> WBCs from the MRV-PCI mandarin fish, respectively. Statistical significance between the control and treated groups is denoted by ****, where the <span class="html-italic">p</span>-value was 0.0001, using a one-way ANOVA and Tukey’s post hoc test.</p>
Full article ">Figure 6
<p>Differential expressions of TLR genes in the WBCs of MRV-PCI mandarin fish following DXMS stimulation. (<b>A</b>) MRV genome DNA copy numbers of the WBCs in the control, 1 day post-treatment and 3 days post-treatment. (<b>B</b>,<b>C</b>) Relative MCP gene and DNA polymerase gene expression in the control, 1 day post-treatment and 3 days post-treatment, respectively. (<b>D</b>) Relative gene expression levels of the 16 TLRs of the WBCs in the control, 1 day post-treatment and 3 days post-treatment. Statistical significance between the control and treated groups is denoted by *, where the <span class="html-italic">p</span>-value was 0.05, denoted by **, where the <span class="html-italic">p</span>-value was 0.01, and denoted by ****, where the <span class="html-italic">p</span>-value was 0.0001, using a one-way ANOVA and Tukey’s post hoc test. n.s., not significant. The gray bars indicate the control group, and the white bars indicate the treatment groups.</p>
Full article ">Figure 7
<p>Assessment of the reactivation of the covert MRV and TLR5 signal by stimulation with <span class="html-italic">E. coli</span> and flagellin. Relative TLR5M gene expression (<b>A</b>), MRV genome copy number (<b>B</b>) and relative MCP expression (<b>C</b>) upon <span class="html-italic">E. coli</span> stimulation. Relative TLR5M gene expression (<b>D</b>), MRV genome copy number (<b>E</b>) and relative MCP expression (<b>F</b>) upon flagellin stimulation. Relative gene expression levels of the WBCs from MRV covert fish stimulated with <span class="html-italic">E. coli</span> and flagellin for 1 or 3 days, or injected with PBS as a control. Statistical significance between the control and treated groups is denoted by**, where the <span class="html-italic">p</span>-value was 0.01 using a one-way ANOVA and Tukey’s post hoc test. n.s, not significant. The gray columns indicate the control group, and the white columns indicate the treatment groups.</p>
Full article ">
Back to TopTop