Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25.483
Filter
1.
Invest Ophthalmol Vis Sci ; 65(8): 1, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949632

ABSTRACT

Purpose: Glucocorticoid-induced glaucoma (GIG) is a prevalent complication associated with glucocorticoids (GCs), resulting in irreversible blindness. GIG is characterized by the abnormal deposition of extracellular matrix (ECM) in the trabecular meshwork (TM), elevation of intraocular pressure (IOP), and loss of retinal ganglion cells (RGCs). The objective of this study is to investigate the effects of nicotinamide riboside (NR) on TM in GIG. Methods: Primary human TM cells (pHTMs) and C57BL/6J mice responsive to GCs were utilized to establish in vitro and in vivo GIG models, respectively. The study assessed the expression of ECM-related proteins in TM and the functions of pHTMs to reflect the effects of NR. Mitochondrial morphology and function were also examined in the GIG cell model. GIG progression was monitored through IOP, RGCs, and mitochondrial morphology. Intracellular nicotinamide adenine dinucleotide (NAD+) levels of pHTMs were enzymatically assayed. Results: NR significantly prevented the expression of ECM-related proteins and alleviated dysfunction in pHTMs after dexamethasone treatment. Importantly, NR protected damaged ATP synthesis, preventing overexpression of mitochondrial reactive oxygen species (ROS), and also protect against decreased mitochondrial membrane potential induced by GCs in vitro. In the GIG mouse model, NR partially prevented the elevation of IOP and the loss of RGCs. Furthermore, NR effectively suppressed the excessive expression of ECM-associated proteins and mitigated mitochondrial damage in vivo. Conclusions: Based on the results, NR effectively enhances intracellular levels of NAD+, thereby mitigating abnormal ECM deposition and TM dysfunction in GIG by attenuating mitochondrial damage induced by GCs. Thus, NR has promising potential as a therapeutic candidate for GIG treatment.


Subject(s)
Disease Models, Animal , Extracellular Matrix , Glaucoma , Glucocorticoids , Intraocular Pressure , Mice, Inbred C57BL , Mitochondria , Niacinamide , Pyridinium Compounds , Trabecular Meshwork , Animals , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Pyridinium Compounds/pharmacology , Glucocorticoids/toxicity , Mitochondria/metabolism , Mitochondria/drug effects , Mice , Glaucoma/metabolism , Glaucoma/drug therapy , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Intraocular Pressure/drug effects , Humans , Trabecular Meshwork/metabolism , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology , Cells, Cultured , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Reactive Oxygen Species/metabolism , Dexamethasone/pharmacology , Male
2.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39000606

ABSTRACT

Sarcopenia refers to an age-related decrease in muscle mass and strength. The gut-muscle axis has been proposed as a promising target to alleviate muscle atrophy. The effect of KL-Biome-a postbiotic preparation comprising heat-killed Lactiplantibacillus plantarum KM-2, its metabolites, and an excipient (soybean powder)-on muscle atrophy was evaluated using dexamethasone (DEX)-induced atrophic C2C12 myoblasts and C57BL/6J mice. KL-Biome significantly downregulated the expression of genes (Atrogin-1 and MuRF1) associated with skeletal muscle degradation but increased the anabolic phosphorylation of FoxO3a, Akt, and mTOR in C2C12 cells. Oral administration of KL-Biome (900 mg/kg) for 8 weeks significantly improved muscle mass, muscle function, and serum lactate dehydrogenase levels in DEX-treated mice. KL-Biome administration increased gut microbiome diversity and reversed DEX-mediated gut microbiota alterations. Furthermore, it significantly increased the relative abundances of the genera Subdologranulum, Alistipes, and Faecalibacterium prausnitzii, which are substantially involved in short-chain fatty acid production. These findings suggest that KL-Biome exerts beneficial effects on muscle atrophy by regulating gut microbiota.


Subject(s)
Dexamethasone , Gastrointestinal Microbiome , Mice, Inbred C57BL , Muscle, Skeletal , Muscular Atrophy , Animals , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Atrophy/chemically induced , Mice , Dexamethasone/pharmacology , Dexamethasone/adverse effects , Gastrointestinal Microbiome/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Male , Muscle Proteins/metabolism , Muscle Proteins/genetics , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Probiotics/administration & dosage , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Sarcopenia/drug therapy , Sarcopenia/metabolism , Sarcopenia/pathology , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Line , Lactobacillus plantarum
3.
Sci Rep ; 14(1): 15304, 2024 07 03.
Article in English | MEDLINE | ID: mdl-38961188

ABSTRACT

Inflammation, corticosteroids, and loading all affect tendon healing, with an interaction between them. However, underlying mechanisms behind the effect of corticosteroids and the interaction with loading remain unclear. The aim of this study was to investigate the role of dexamethasone during tendon healing, including specific effects on tendon cells. Rats (n = 36) were randomized to heavy loading or mild loading, the Achilles tendon was transected, and animals were treated with dexamethasone or saline. Gene and protein analyses of the healing tendon were performed for extracellular matrix-, inflammation-, and tendon cell markers. We further tested specific effects of dexamethasone on tendon cells in vitro. Dexamethasone increased mRNA levels of S100A4 and decreased levels of ACTA2/α-SMA, irrespective of load level. Heavy loading + dexamethasone reduced mRNA levels of FN1 and TenC (p < 0.05), while resolution-related genes were unaltered (p > 0.05). In contrast, mild loading + dexamethasone increased mRNA levels of resolution-related genes ANXA1, MRC1, PDPN, and PTGES (p < 0.03). Altered protein levels were confirmed in tendons with mild loading. Dexamethasone treatment in vitro prevented tendon construct formation, increased mRNA levels of S100A4 and decreased levels of SCX and collagens. Dexamethasone during tendon healing appears to act through immunomodulation by promoting resolution, but also through an effect on tendon cells.


Subject(s)
Achilles Tendon , Dexamethasone , Tendon Injuries , Wound Healing , Dexamethasone/pharmacology , Animals , Rats , Wound Healing/drug effects , Tendon Injuries/drug therapy , Tendon Injuries/metabolism , Achilles Tendon/drug effects , Achilles Tendon/metabolism , Achilles Tendon/injuries , Achilles Tendon/pathology , S100 Calcium-Binding Protein A4/metabolism , S100 Calcium-Binding Protein A4/genetics , Male , Annexin A1/metabolism , Annexin A1/genetics , Actins/metabolism , Actins/genetics , Collagen/metabolism , Rats, Sprague-Dawley , Tendons/drug effects , Tendons/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , RNA, Messenger/metabolism , RNA, Messenger/genetics , Basic Helix-Loop-Helix Transcription Factors
4.
Sci Rep ; 14(1): 15349, 2024 07 03.
Article in English | MEDLINE | ID: mdl-38961190

ABSTRACT

Precision-cut liver slices (PCLS) are increasingly used as a model to investigate anti-fibrotic therapies. However, many studies use PCLS from healthy animals treated with pro-fibrotic stimuli in culture, which reflects only the early stages of fibrosis. The effects of different culture conditions on PCLS from cirrhotic animals has not been well characterized and there is no consensus on optimal methods. In this study, we report a method for the collection and culture of cirrhotic PCLS and compare the effect of common culture conditions on viability, function, and gene expression. Additionally, we compared three methods of RNA isolation and identified a protocol with high yield and purity. We observed significantly increased albumin production when cultured with insulin-transferrin-selenium and dexamethasone, and when incubated on a rocking platform. Culturing with insulin-transferrin-selenium and dexamethasone maintained gene expression closer to the levels in fresh slices. However, despite stable viability and function up to 4 days, we found significant changes in expression of key genes by day 2. Interestingly, we also observed that cirrhotic PCLS maintain viability in culture longer than slices from healthy animals. Due to the influence of matrix stiffness on fibrosis and hepatocellular function, it is important to evaluate prospective anti-fibrotic therapies in a platform that preserves tissue biomechanics. PCLS from cirrhotic animals represent a promising tool for the development of treatments for chronic liver disease.


Subject(s)
Dexamethasone , Liver Cirrhosis , Liver , Animals , Rats , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Dexamethasone/pharmacology , Male , RNA/isolation & purification , RNA/genetics , RNA/metabolism , Insulin/metabolism , Insulin/pharmacology , Rats, Sprague-Dawley , Selenium/pharmacology , Tissue Culture Techniques/methods
5.
Int J Med Sci ; 21(8): 1511-1517, 2024.
Article in English | MEDLINE | ID: mdl-38903930

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSCs), which are capable of differentiating into osteoblasts, are used in effective regenerative therapies. MSCs must be prompted to differentiate into osteoblasts for MSC transplantation to be effective. In this study, osteoblast differentiation markers involved in bone formation were evaluated to investigate the stress resistance of bone marrow-derived rat MSCs to dexamethasone and hypoxia and their ability to differentiate into osteoblasts. MSCs were allowed to differentiate into osteoblasts for 21 days in three different environments (dexamethasone treatment, hypoxic conditions [1% oxygen], or both). Osteoblast differentiation potential was evaluated according to alkaline phosphatase levels and a mineralisation assay. Immunofluorescence staining was used to determine the protein expression of the osteoblast differentiation markers type I collagen and osteopontin. MSCs differentiated into osteoblasts under hypoxic conditions but differentiated more slowly upon treatment with dexamethasone and dexamethasone plus hypoxia relative to the control. MSCs preconditioned with dexamethasone or hypoxia and then allowed to differentiate into osteoblasts under similar conditions differentiated comparably to control MSCs. MSCs that developed resistance to dexamethasone or hypoxia differentiated more quickly into osteoblasts than those that did not. The findings suggest that increasing the resistance of MSCs to stress by preconditioning them via dexamethasone or hypoxia exposure could result in more rapid differentiation into osteoblasts following transplantation.


Subject(s)
Cell Differentiation , Cell Hypoxia , Dexamethasone , Mesenchymal Stem Cells , Osteoblasts , Dexamethasone/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/metabolism , Cell Differentiation/drug effects , Rats , Cell Hypoxia/drug effects , Osteogenesis/drug effects , Cells, Cultured , Alkaline Phosphatase/metabolism , Humans , Mesenchymal Stem Cell Transplantation/methods , Collagen Type I/metabolism , Male
6.
Biochem Biophys Res Commun ; 723: 150188, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38824808

ABSTRACT

Steroid (glucocorticoid)-induced necrosis of the femoral head (SONFH) represents a prevalent, progressive, and challenging bone and joint disease characterized by diminished osteogenesis and angiogenesis. Omaveloxolone (OMA), a semi-synthetic oleanocarpane triterpenoid with antioxidant, anti-inflammatory, and osteogenic properties, emerges as a potential therapeutic agent for SONFH. This study investigates the therapeutic impact of OMA on SONFH and elucidates its underlying mechanism. The in vitro environment of SONFH cells was simulated by inducing human bone marrow mesenchymal stem cells (hBMSCs) and human umbilical vein endothelial cells (HUVECs) using dexamethasone (DEX).Various assays, including CCK-8, alizarin red staining, Western blot, qPCR, immunofluorescence, flow cytometry, and TUNNEL, were employed to assess cell viability, STING/NF-κB signaling pathway-related proteins, hBMSCs osteogenesis, HUVECs migration, angiogenesis, and apoptosis. The results demonstrate that OMA promotes DEX-induced osteogenesis, HUVECs migration, angiogenesis, and anti-apoptosis in hBMSCs by inhibiting the STING/NF-κB signaling pathway. This experimental evidence underscores the potential of OMA in regulating DEX-induced osteogenesis, HUVECs migration, angiogenesis, and anti-apoptosis in hBMSCs through the STING/NF-κB pathway, thereby offering a promising avenue for improving the progression of SONFH.


Subject(s)
Femur Head Necrosis , Glucocorticoids , Human Umbilical Vein Endothelial Cells , Neovascularization, Physiologic , Osteogenesis , Humans , Osteogenesis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Femur Head Necrosis/chemically induced , Femur Head Necrosis/pathology , Femur Head Necrosis/drug therapy , Femur Head Necrosis/metabolism , Neovascularization, Physiologic/drug effects , Glucocorticoids/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cells, Cultured , Apoptosis/drug effects , Dexamethasone/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Femur Head/pathology , Femur Head/drug effects , Femur Head/blood supply , Femur Head/metabolism , Cell Movement/drug effects , Triterpenes/pharmacology , Cell Survival/drug effects , Angiogenesis
7.
J Nutr Sci Vitaminol (Tokyo) ; 70(3): 219-227, 2024.
Article in English | MEDLINE | ID: mdl-38945887

ABSTRACT

This study investigated the protective effect of carnosine and its components (L-histidine and ß-alanine [HA]) against dexamethasone (Dex)-induced muscle atrophy in C2C12 myotubes. Myotubes were treated with Dex (10 µM) to induce muscle atrophy manifested by decreased myotube diameter, low myosin heavy chain content, and increased expression of muscle atrophy-associated ubiquitin ligases (Atrogin-1, MuRF-1, and Cbl-b). Carnosine (20 mM) treatment significantly improved the myotube diameter and MyHC protein expression level in Dex-treated C2C12 myotubes. It also downregulated the expression of Atrogin-1, MuRF-1, and Cbl-b and suppressed the expression of forkhead box O3 (FoxO3a) mediated by Dex. Furthermore, reactive oxygen species production was increased by Dex but was ameliorated by carnosine treatment. However, HA (20 mM), the component of carnosine, treatment was found ineffective in preventing Dex-induced protein damage. Therefore, based on above results it can be suggested that carnosine could be a potential therapeutic agent to prevent Dex-induced muscle atrophy compared to its components HA.


Subject(s)
Carnosine , Dexamethasone , Muscle Fibers, Skeletal , Muscle Proteins , Muscular Atrophy , Reactive Oxygen Species , SKP Cullin F-Box Protein Ligases , Carnosine/pharmacology , Dexamethasone/pharmacology , Muscular Atrophy/chemically induced , Muscular Atrophy/prevention & control , Muscular Atrophy/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Animals , Mice , Muscle Proteins/metabolism , Cell Line , Reactive Oxygen Species/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism , Forkhead Box Protein O3/metabolism , Tripartite Motif Proteins/metabolism , Myosin Heavy Chains/metabolism
8.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38928510

ABSTRACT

The decline in the function and mass of skeletal muscle during aging or other pathological conditions increases the incidence of aging-related secondary diseases, ultimately contributing to a decreased lifespan and quality of life. Much effort has been made to surmise the molecular mechanisms underlying muscle atrophy and develop tools for improving muscle function. Enhancing mitochondrial function is considered critical for increasing muscle function and health. This study is aimed at evaluating the effect of an aqueous extract of Gloiopeltis tenax (GTAE) on myogenesis and muscle atrophy caused by dexamethasone (DEX). The GTAE promoted myogenic differentiation, accompanied by an increase in peroxisome proliferator-activated receptor γ coactivator α (PGC-1α) expression and mitochondrial content in myoblast cell culture. In addition, the GTAE alleviated the DEX-mediated myotube atrophy that is attributable to the Akt-mediated inhibition of the Atrogin/MuRF1 pathway. Furthermore, an in vivo study using a DEX-induced muscle atrophy mouse model demonstrated the efficacy of GTAE in protecting muscles from atrophy and enhancing mitochondrial biogenesis and function, even under conditions of atrophy. Taken together, this study suggests that the GTAE shows propitious potential as a nutraceutical for enhancing muscle function and preventing muscle wasting.


Subject(s)
Dexamethasone , Muscle Development , Muscular Atrophy , Plant Extracts , Animals , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , Muscular Atrophy/drug therapy , Muscular Atrophy/pathology , Dexamethasone/adverse effects , Dexamethasone/pharmacology , Muscle Development/drug effects , Mice , Plant Extracts/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Cell Differentiation/drug effects , Myoblasts/drug effects , Myoblasts/metabolism , Cell Line , Muscle Proteins/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Mice, Inbred C57BL , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Rhodophyta
9.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38928138

ABSTRACT

Based on the lack of differences in progression-free and overall survival after a median follow-up of 93 months in our HOVON-65/GMMG-HD4 trial (German part; n = 395) randomizing VAD induction (vincristin/adriamycin/dexamthasone)/tandem-transplantation/thalidomide-maintenance vs. PAD induction (bortezomib/adriamycin/dexamethasone)/tandem transplantation/bortezomib maintenance, we discern how chromosomal aberrations determine long-term prognosis by different patterns of association with proliferation and treatment-dependent response, whether responses achieved by different regimens are equal regarding prognosis, and whether subpopulations of patients could be defined as treatable without upfront "novel agents" in cases of limited resources, e.g., in low- or middle-income countries. Serum parameters and risk factors were assessed in 395 patients. CD138-purified plasma cells were subjected to fluorescence in situ hybridization (n = 354) and gene expression profiling (n = 204). We found chromosomal aberrations to be associated in four patterns with survival, proliferation, and response: deletion (del) del17p13, del8p21, del13q14, (gain) 1q21+, and translocation t(4;14) (all adverse) associate with higher proliferation. Of these, del17p is associated with an adverse response (pattern 1), and 1q21+, t(4;14), and del13q14 with a treatment-dependent better response (pattern 2). Hyperdiploidy associates with lower proliferation without impacting response or survival (pattern 3). Translocation t(11;14) has no association with survival but a treatment-dependent adverse response (pattern 4). Significantly fewer patients reach a near-complete response or better with "conventional" (VAD) vs. bortezomib-based treatment after induction or high-dose melphalan. These patients, however, show significantly better median progression-free and overall survival. Molecularly, patients responding to the two regimens differ in gene expression, indicating distinct biological properties of the responding myeloma cells. Patients with normal renal function (89.4%), low cytogenetic risk (72.5%), or low proliferation rate (37.9%) neither benefit in progression-free nor overall survival from bortezomib-based upfront treatment. We conclude that response level, the treatment by which it is achieved, and molecular background determine long-term prognosis. Chromosomal aberrations are associated in four patterns with proliferation and treatment-dependent responses. Associations with faster and deeper responses can be deceptive in the case of prognostically adverse aberrations 1q21+ and t(4;14). Far from advocating a return to "outdated" treatments, if resources do not permit state-of-the-art-treatment, normal renal function and/or molecular profiling identifies patient subpopulations doing well without upfront "novel agents".


Subject(s)
Chromosome Aberrations , Multiple Myeloma , Humans , Multiple Myeloma/genetics , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Female , Male , Middle Aged , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation/drug effects , Prognosis , Adult , Developing Countries , Dexamethasone/therapeutic use , Dexamethasone/pharmacology , Bortezomib/therapeutic use , Bortezomib/pharmacology , Thalidomide/therapeutic use
10.
Int Immunopharmacol ; 136: 112395, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38833845

ABSTRACT

Asthma is a long-term disease that causes airways swelling and inflammation and in turn airway narrowing. AdipoRonis an orally active synthetic small molecule that acts as a selective agonist at theadiponectin receptor 1 and 2. The aim of the current study is to delineate the protective effect and the potential underlying mechanism ofadipoRon inairway inflammationinduced byovalbumin (OVA) in comparison withdexamethasone. Adult maleSwiss Albino micewere sensitized to OVA on days 0 and 7, then challenged with OVA on days 14, 15 and 16. AdipoRon was administered orally for 6 days starting from the 11th day till the 16th and 1 h prior to OVA in the challenge days. Obtained results from asthmatic control group showed a significant decrease in serum adiponectin concentration, an increase in inflammatory cell counts inthe bronchoalveolar lavage fluid(BALF), CD68 protein expression, inflammatory cytokine concentration and oxidative stress as well. Administration of adipoRon enhanced antioxidant mechanisms limiting oxidative stress by significantly increasing reduced glutathione (GSH) pulmonary content, decreasing serum lactate dehydrogenase (LDH) together with malondialdehyde (MDA) significant reduction in lung tissue. In addition, it modulated the levels of serum immunoglobulin E (IgE), pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-4, IL-13, nuclear factor kappa B (NF-κB) and the anti-inflammatory one IL-10 improving lung inflammation as revealed by histopathological evaluation. Furthermore, lung tissue expression of nuclear factor erythroid 2-related factor (Nrf2) and 5'AMP-activated protein kinase (AMPK) were significantly increased adipoRon. Notably, results of adipoRon received group were comparable to those of dexamethasone group. In conclusion, our study demonstrates that adipoRon can positively modulate adiponectin expression with activation of AMPK pathway and subsequent improvement in inflammatory and oxidative signaling.


Subject(s)
AMP-Activated Protein Kinases , Asthma , Disease Models, Animal , Ovalbumin , Receptors, Adiponectin , Signal Transduction , Animals , Asthma/drug therapy , Asthma/immunology , Asthma/chemically induced , Asthma/metabolism , Mice , Receptors, Adiponectin/agonists , Receptors, Adiponectin/metabolism , Ovalbumin/immunology , Male , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Lung/pathology , Lung/drug effects , Lung/immunology , Cytokines/metabolism , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Oxidative Stress/drug effects , Adiponectin , Anti-Asthmatic Agents/therapeutic use , Anti-Asthmatic Agents/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Immunoglobulin E/blood , Humans , Dexamethasone/therapeutic use , Dexamethasone/pharmacology , Piperidines
11.
Drug Des Devel Ther ; 18: 2043-2061, 2024.
Article in English | MEDLINE | ID: mdl-38863767

ABSTRACT

Background: Despite its extensive utilization in Chinese hospitals for treating acute pancreatitis (AP) and related acute respiratory distress syndrome (ARDS), the active components and mechanisms underlying the action of Qingyi Granule (QYKL) remain elusive. Methods: This study consists of four parts. First, we used Mendelian randomization (MR) to investigate the causal relationship between AP, cytokine, and ARDS. Next, 321 patients were collected to evaluate the efficacy of QYKL combined with dexamethasone (DEX) in treating AP. In addition, we used UHPLC-QE-MS to determine the chemical constituents of QYKL extract and rat serum after the oral administration of QYKL. The weighted gene coexpression network analysis (WGCNA) method was used to find the main targets of AP-related ARDS using the GSE151572 dataset. At last, a AP model was established by retrograde injection of 5% sodium taurocholate. Results: MR showed that AP may have a causal relationship with ARDS by mediating cytokine storms. Retrospective study results showed early administration of QYKL was associated with a lower incidence of ARDS, mortality, admissions to the intensive care unit, and length of stay in AP patients compared to the Control group. Furthermore, we identified 23 QYKL prototype components absorbed into rat serum. WGCNA and differential expression analysis identified 1558 APALI-related genes. The prototype components exhibited strong binding activity with critical targets. QYKL has a significant protective effect on pancreatic and lung injury in AP rats, and the effect is more effective after combined treatment with DEX, which may be related to the regulation of the IL-6/STAT3 signaling pathway. Conclusion: By integrating MR, retrospective analysis, and systematic pharmacological methodologies, this study systematically elucidated the therapeutic efficacy of QYKL in treating AP-related ARDS, establishing a solid foundation for its medicinal use.


Subject(s)
Drugs, Chinese Herbal , Pancreatitis , Respiratory Distress Syndrome , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Respiratory Distress Syndrome/drug therapy , Pancreatitis/drug therapy , Pancreatitis/metabolism , Animals , Rats , Humans , Retrospective Studies , Male , Rats, Sprague-Dawley , Dexamethasone/pharmacology , Dexamethasone/administration & dosage , Acute Disease , Female , Middle Aged
12.
Res Vet Sci ; 175: 105318, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38851053

ABSTRACT

A retrospective cross-sectional study was conducted to assess the frequency of low-dose dexamethasone suppression test (LDDST) patterns in canine patients that had clinicopathologic signs consistent with Cushing's syndrome (CS). Medical records for patients of interest (N = 128) were reviewed between January 2014 and December 2020 to analyse and classify LDDST results based upon the following patterns: lack of suppression, partial suppression, complete suppression, escape, or inverse. Complete suppression, lack of suppression, partial suppression, escape, and inverse patterns were identified in 39.1%, 31.2%, 14.1%, 10.1% and 5.5% of cases respectively. LDDST results were also evaluated with respect to clinical signs, serum alkaline phosphatase (ALP) activity, urine specific gravity (USG) and adrenal ultrasonographic findings. There was no association between LDDST patterns and clinical signs (p = 0.11), increased ALP (p = 0.32), USG (p = 0.33) or adrenal ultrasonographic findings (p = 0.19). In all dogs that demonstrated complete suppression or an inverse pattern, CS was excluded by the attending clinician. The diagnosis of CS was also excluded without further exploration in 23.1%, 7.5% and 5.6% of dogs that demonstrated an escape pattern, lack of suppression and partial suppression pattern, respectively. These results suggest that the clinical significance of LDDST patterns, particularly escape and inverse patterns, are misunderstood by some clinicians, leading them to prematurely exclude the diagnosis of CS.


Subject(s)
Cushing Syndrome , Dexamethasone , Dog Diseases , Dogs , Animals , Retrospective Studies , Dog Diseases/diagnostic imaging , Cushing Syndrome/veterinary , Cushing Syndrome/pathology , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Male , Cross-Sectional Studies , Female , Ultrasonography/veterinary
13.
Int Immunopharmacol ; 137: 112519, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38901241

ABSTRACT

PURPOSE: Patients with nanophthalmos might be prone to developing intraocular inflammation following an acute glaucoma attack. Here, we aimed to investigate the role of MYRF in intraocular inflammation by modeling the mutation in mice. METHODS: Nanophthalmos frameshift mutation of Myrf was introduced into the mouse genome with the CRISPR-Cas9 system. Signaling pathways in eye tissues were delineated using RNA sequencing and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Intraocular inflammation was induced by a lipopolysaccharide (LPS) intravitreal injection. Dexamethasone (DEX) was administered systemically and locally a week before the LPS injection. The anterior segment clinical scores of the mice were examined 24 h after the LPS injection. Infiltrating inflammatory cells were evaluated with histopathology and immunofluorescence. The mRNA levels of inflammatory cytokines were quantified with reverse transcription-quantitative PCR (RT-qPCR) and the corresponding protein concentrations using enzyme-linked immunosorbent assay (ELISA). RESULTS: Many inflammation-associated signaling pathways were enriched in Myrf mut/+ mice ocular tissues. Clinical scores of Myrf mut/+ mice were significantly higher than those of Myrf +/+ mice 24 h after LPS administration. Histological examination demonstrated high inflammatory cell infiltration in the anterior and vitreous chambers in Myrf mut/+ mice, with numerous CD45+ and CD11b+ inflammatory cells. Moreover, enhanced expression of inflammatory cytokines MCP-1, TGF-ß, and IL-1ß in eyes and aqueous humor of Myrf mut/+ mice was detected. Remarkably, pretreating Myrf mut/+ mice with DEX relieved the intraocular inflammation. CONCLUSION: Nanophthalmos-associated MYRF mutation renders mouse eyes more susceptible to inflammation. Dexamethasone treatment ameliorates the inflammatory response.


Subject(s)
Cytokines , Dexamethasone , Lipopolysaccharides , Microphthalmos , Animals , Mice , Dexamethasone/therapeutic use , Dexamethasone/pharmacology , Microphthalmos/genetics , Cytokines/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Inflammation/genetics , Signal Transduction , Frameshift Mutation , Male , Humans
14.
Nat Commun ; 15(1): 5483, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38942804

ABSTRACT

Dexamethasone is the standard of care for critically ill patients with COVID-19, but the mechanisms by which it decreases mortality and its immunological effects in this setting are not understood. Here we perform bulk and single-cell RNA sequencing of samples from the lower respiratory tract and blood, and assess plasma cytokine profiling to study the effects of dexamethasone on both systemic and pulmonary immune cell compartments. In blood samples, dexamethasone is associated with decreased expression of genes associated with T cell activation, including TNFSFR4 and IL21R. We also identify decreased expression of several immune pathways, including major histocompatibility complex-II signaling, selectin P ligand signaling, and T cell recruitment by intercellular adhesion molecule and integrin activation, suggesting these are potential mechanisms of the therapeutic benefit of steroids in COVID-19. We identify additional compartment- and cell- specific differences in the effect of dexamethasone that are reproducible in publicly available datasets, including steroid-resistant interferon pathway expression in the respiratory tract, which may be additional therapeutic targets. In summary, we demonstrate compartment-specific effects of dexamethasone in critically ill COVID-19 patients, providing mechanistic insights with potential therapeutic relevance. Our results highlight the importance of studying compartmentalized inflammation in critically ill patients.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Cytokines , Dexamethasone , Lung , SARS-CoV-2 , Dexamethasone/therapeutic use , Dexamethasone/pharmacology , Humans , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/drug effects , Lung/drug effects , Lung/virology , Cytokines/metabolism , Cytokines/blood , Critical Illness , Male , Single-Cell Analysis , Female , Middle Aged , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Aged , Lymphocyte Activation/drug effects
15.
Biosci Rep ; 44(7)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38864530

ABSTRACT

Tamoxifen (TAM) is a key player in estrogen receptor-positive (ER+) breast cancer (BC); however, ∼30% of patients experience relapse and a lower survival rate due to TAM resistance. TAM resistance was related to the over expression of SOX-2 gene, which is regulated by the E2F3 transcription factor in the Wnt signaling pathway. It was suggested that SOX-2 overexpression was suppressed by dexamethasone (DEX), a glucocorticoid commonly prescribed to BC patients. The aim of the present study is to explore the effect of combining DEX and TAM on the inhibition of TAM-resistant LCC-2 cells (TAMR-1) through modulating the E2F3/SOX-2-mediated Wnt signaling pathway. The effect of the combination therapy on MCF-7 and TAMR-1 cell viability was assessed. Drug interactions were analyzed using CompuSyn and SynergyFinder softwares. Cell cycle distribution, apoptotic protein expression, gene expression levels of SOX-2 and E2F3, and cell migration were also assessed. Combining DEX with TAM led to synergistic inhibition of TAMR-1 cell proliferation and migration, induced apoptosis, reduced SOX-2 and E2F3 expression and was also associated with S and G2-M phase arrest. Therefore, combining DEX with TAM may present an effective therapeutic option to overcome TAM resistance, by targeting the E2F3/SOX-2/Wnt signaling pathway, in addition to its anti-inflammatory effect.


Subject(s)
Breast Neoplasms , Cell Proliferation , Dexamethasone , Drug Resistance, Neoplasm , Drug Synergism , Tamoxifen , Humans , Tamoxifen/pharmacology , Dexamethasone/pharmacology , Drug Resistance, Neoplasm/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Female , MCF-7 Cells , Cell Proliferation/drug effects , Apoptosis/drug effects , Cell Movement/drug effects , Wnt Signaling Pathway/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , E2F3 Transcription Factor/metabolism , E2F3 Transcription Factor/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics
16.
Iran J Allergy Asthma Immunol ; 23(2): 197-220, 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38822514

ABSTRACT

Systemic sclerosis (SSc) is an autoimmune systemic disease that is characterized by immune dysregulation, inflammation, vasculopathy, and fibrosis. Tissue fibrosis plays an important role in SSc and can affect several organs such as the dermis, lungs, and heart. Dysregulation of interferon (IFN) signaling contributes to the SSc pathogenesis and interferon regulatory factor 1 (IRF1) has been indicated as the main regulator of type I IFN. This study aimed to clarify the effect of IFN-gamma (-γ) and dexamethasone (DEX) on the IRF1, extracellular signal-regulated kinase 1/2 (ERK1/2), and the expression of alpha-smooth muscle actin (α-SMA) in myofibroblasts and genes involved in the inflammation and fibrosis processes in early diffuse cutaneous systemic sclerosis (dcSSc). A total of 10 early dcSSc patients (diffuse cutaneous form) and 10 unaffected control dermis biopsies were obtained to determine IFNγ and DEX effects on inflammation and fibrosis. Fibroblasts were treated with IFNγ and DEX at optimum time and dose. The expression level of genes and proteins involved in the fibrosis and inflammation processes have been quantified by quantitative real-time PCR (RT-qPCR) and western blot, respectively. IFNγ could up-regulate some of the inflammation-related genes (Interleukin-6; IL6) and down-regulate some of the fibrosis-related genes (COL1A1) in cultured fibroblasts of patients with early dcSSc compared to the untreated group. Besides, it has been revealed that IFNγ can induce fibroblast differentiation to the myofibroblast that expresses α-SMA. Concerning the inhibitory effect of IFNγ on some fibrotic genes and its positive effect on the inflammatory genes and myofibroblast differentiation, it seems that IFNγ may play a dual role in SSc.


Subject(s)
Actins , Fibroblasts , Interferon-gamma , Interleukin-6 , Scleroderma, Systemic , Adult , Female , Humans , Male , Middle Aged , Actins/metabolism , Actins/genetics , Cells, Cultured , Dexamethasone/pharmacology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Interleukin-6/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/immunology
17.
Stress ; 27(1): 2353781, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38823417

ABSTRACT

Hypothalamic-pituitary-adrenal (HPA)-axis hyperactivity measured by the combined dexamethasone-CRH test (DEX-CRH test) has been found in patients with major depressive disorder (MDD), whereas hypoactivity has been found in patients with work-related stress. We aimed to investigate the DEX-CRH test as a biomarker to distinguish between MDD and work-related stress (exhaustion disorder - ED). We hypothesized that there would be lower cortisol and ACTH response in participants with ED compared to MDD and healthy controls (HC). Also, we explored if the cortisol response of those patients interacted with robust markers of oxidative stress. Thirty inpatients with MDD and 23 outpatients with ED were recruited. Plasma cortisol and ACTH were sampled during a DEX-CRH test. The main outcome measure, area under the curve (AUC) for cortisol and ACTH, was compa-red between MDD vs. ED participants and a historical HC group. Secondary markers of oxidative stress urinary 8-oxodG and 8-oxoGuo; quality of sleep and psychometrics were obtained. Cortisol concentrations were higher in MDD and ED participants compared to HC, and no differences in AUC cortisol and ACTH were found between ED vs. MDD. Compared to ED, MDD participants had higher stress symptom severity and a lower sense of well-being. No differences in oxidative stress markers or quality of sleep between the groups were found. The result indicates that the patients with ED, like patients with MDD, are non-suppressors in DEX-CRH test and not hypocortisolemic as suggested.


Subject(s)
Adrenocorticotropic Hormone , Biomarkers , Depressive Disorder, Major , Dexamethasone , Hydrocortisone , Oxidative Stress , Humans , Depressive Disorder, Major/blood , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/diagnosis , Female , Male , Hydrocortisone/blood , Adult , Oxidative Stress/physiology , Adrenocorticotropic Hormone/blood , Biomarkers/blood , Dexamethasone/pharmacology , Middle Aged , Corticotropin-Releasing Hormone/blood , Occupational Stress/physiopathology , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/physiopathology
18.
Exp Cell Res ; 440(1): 114137, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38897410

ABSTRACT

Glaucoma is characterized by pathological elevation of intraocular pressure (IOP) due to dysfunctional trabecular meshwork (TM), which is the primary cause of irreversible vision loss. There are currently no effective treatment strategies for glaucoma. Mitochondrial function plays a crucial role in regulating IOP within the TM. In this study, primary TM cells treated with dexamethasone were used to simulate glaucomatous changes, showing abnormal cellular cytoskeleton, increased expression of extracellular matrix, and disrupted mitochondrial fusion and fission dynamics. Furthermore, glaucomatous TM cell line GTM3 exhibited impaired mitochondrial membrane potential and phagocytic function, accompanied by decreased oxidative respiratory levels as compared to normal TM cells iHTM. Mechanistically, lower NAD + levels in GTM3, possibly associated with increased expression of key enzymes CD38 and PARP1 related to NAD + consumption, were observed. Supplementation of NAD + restored mitochondrial function and cellular viability in GTM3 cells. Therefore, we propose that the aberrant mitochondrial function in glaucomatous TM cells may be attributed to increased NAD + consumption dependent on CD38 and PARP1, and NAD + supplementation could effectively ameliorate mitochondrial function and improve TM function, providing a novel alternative approach for glaucoma treatment.


Subject(s)
Glaucoma , Mitochondria , NAD , Trabecular Meshwork , Trabecular Meshwork/metabolism , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/drug therapy , NAD/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Intraocular Pressure/drug effects , Cell Survival/drug effects , ADP-ribosyl Cyclase 1/metabolism , ADP-ribosyl Cyclase 1/genetics , Cell Line , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Dexamethasone/pharmacology , Cells, Cultured
19.
Immunology ; 172(4): 653-668, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38786548

ABSTRACT

The mechanisms underlying the development of steroid resistance in asthma remain unclear. To establish whether as well as the mechanisms by which the activation of Janus kinases (JAKs) is involved in the development of steroid resistance in asthma, murine steroid-resistant models of the proliferation of group 2 innate lymphoid cells (ILC2s) in vitro and asthmatic airway inflammation in vivo were analysed. ILC2s in the lungs of BALB/c mice were sorted and then incubated with IL-33, thymic stromal lymphopoietin (TSLP), and/or IL-7 with or without dexamethasone (10 nM), the pan-JAK inhibitor, delgocitinib (1-10 000 nM), and/or the Bcl-xL inhibitor, navitoclax (1-100 nM), followed by the detection of viable and apoptotic cells. The anti-apoptotic factor, Bcl-xL was detected in ILC2s by flow cytometry. As a steroid-resistant asthma model, ovalbumin (OVA)-sensitized BALB/c mice were intratracheally challenged with OVA at a high dose of 500 µg four times. Dexamethasone (1 mg/kg, i.p.), delgocitinib (3-30 mg/kg, p.o.), or navitoclax (30 mg/kg, p.o.) was administered during the challenges. Cellular infiltration into the lungs was analysed by flow cytometry. Airway remodelling was histologically evaluated. The following results were obtained. (1) Cell proliferation concomitant with a decrease in apoptotic cells was induced when ILC2s were cultured with TSLP and/or IL-7, and was potently inhibited by dexamethasone. In contrast, when the culture with TSLP and IL-7 was performed in the presence of IL-33, the proliferative response exhibited steroid resistance. Steroid-resistant ILC2 proliferation was suppressed by delgocitinib in a concentration-dependent manner. (2) The culture with IL-33, TSLP, and IL-7 induced the overexpression of Bcl-xL, which was clearly inhibited by delgocitinib, but not by dexamethasone. When ILC2s were treated with navitoclax, insensitivity to dexamethasone was significantly cancelled. (3) The development of airway remodelling and the infiltration of ILC2s into the lungs in the asthma model were not suppressed by dexamethasone, but were dose-dependently inhibited by delgocitinib. Combination treatment with dexamethasone and either delgocitinib or navitoclax synergistically suppressed these responses. Therefore, JAKs appear to play significant roles in the induction of steroid resistance by up-regulating Bcl-xL in ILC2s. The inhibition of JAKs and Bcl-xL has potential as pharmacotherapy for steroid-resistant asthma, particularly that mediated by ILC2s.


Subject(s)
Asthma , Dexamethasone , Drug Resistance , Immunity, Innate , Janus Kinases , Lymphocytes , Mice, Inbred BALB C , bcl-X Protein , Animals , Asthma/drug therapy , Asthma/immunology , Asthma/metabolism , bcl-X Protein/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/drug effects , Mice , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Immunity, Innate/drug effects , Janus Kinases/metabolism , Lung/immunology , Lung/pathology , Lung/drug effects , Female , Cytokines/metabolism , Disease Models, Animal , Apoptosis/drug effects , Cell Proliferation/drug effects , Interleukin-33/metabolism , Thymic Stromal Lymphopoietin , Sulfonamides/pharmacology
20.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 712-719, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708505

ABSTRACT

OBJECTIVE: To explore the mechanism underlying the protective effect of α2-macroglobulin (A2M) against glucocorticoid-induced femoral head necrosis. METHODS: In a human umbilical vein endothelial cell (HUVEC) model with injuries induced by gradient concentrations of dexamethasone (DEX; 10-8-10-5 mol/L), the protective effects of A2M at 0.05 and 0.1 mg/mL were assessed by examining the changes in cell viability, migration, and capacity of angiogenesis using CCK-8 assay, Transwell and scratch healing assays and angiogenesis assay. The expressions of CD31 and VEGF-A proteins in the treated cells were detected using Western blotting. In BALB/c mouse models of avascular necrosis of the femoral head induced by intramuscular injections of methylprednisolone, the effects of intervention with A2M on femoral trabecular structure, histopathological characteristics, and CD31 expression were examined with Micro-CT, HE staining and immunohistochemical staining. RESULTS: In cultured HUVECs, DEX treatment significantly reduced cell viability, migration and angiogenic ability in a concentration- and time-dependent manner (P<0.05), and these changes were obviously reversed by treatment with A2M in positive correlation with A2M concentration (P<0.05). DEX significantly reduced the expression of CD31 and VEGF-A proteins in HUVECs, while treatment with A2M restored CD31 and VEGF-A expressions in the cells (P<0.05). The mouse models of femoral head necrosis showed obvious trabecular damages in the femoral head, where a large number of empty lacunae and hypertrophic fat cells could be seen and CD31 expression was significantly decreased (P<0.05). A2M treatment of the mouse models significantly improved trabecular damages, maintained normal bone tissue structures, and increased CD31 expression in the femoral head (P<0.05). CONCLUSION: A2M promotes proliferation, migration, and angiogenesis of DEX-treated HUVECs and alleviates methylprednisolone-induced femoral head necrosis by improving microcirculation damages and maintaining microcirculation stability in the femoral head.


Subject(s)
Cell Movement , Cell Proliferation , Dexamethasone , Femur Head Necrosis , Glucocorticoids , Human Umbilical Vein Endothelial Cells , Mice, Inbred BALB C , Animals , Mice , Femur Head Necrosis/chemically induced , Femur Head Necrosis/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Glucocorticoids/adverse effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Dexamethasone/adverse effects , Dexamethasone/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Cell Survival/drug effects , Femur Head/pathology , Femur Head/blood supply , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Angiogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...