Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 65.274
1.
Molecules ; 29(9)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38731499

Carbon nanodots (CDs) are commonly found in food products and have attracted significant attention from food scientists. There is a high probability of CD exposure in humans, but its impacts on health are unclear. Therefore, health effects associated with CD consumption should be investigated. In this study, we attempted to create a model system of the Maillard reaction between cystine and glucose using a simple cooking approach. The CDs (CG-CDs) were isolated from cystine-glucose-based Maillard reaction products and characterized using fluorescence spectroscopy, X-ray diffractometer (XRD), and transmission electron microscope (TEM). Furthermore, human mesenchymal stem cells (hMCs) were used as a model to unravel the CDs' cytotoxic properties. The physiochemical assessment revealed that CG-CDs emit excitation-dependent fluorescence and possess a circular shape with sizes ranging from 2 to 13 nm. CG-CDs are predominantly composed of carbon, oxygen, and sulfur. The results of the cytotoxicity evaluation indicate good biocompatibility, where no severe toxicity was observed in hMCs up to 400 µg/mL. The DPPH assay demonstrated that CDs exert potent antioxidant abilities. The qPCR analysis revealed that CDs promote the downregulation of the key regulatory genes, PPARγ, C/EBPα, SREBP-1, and HMGCR, coupled with the upregulation of anti-inflammatory genes. Our findings suggested that, along with their excellent biocompatibility, CG-CDs may offer positive health outcomes by modulating critical genes involved in lipogenesis, homeostasis, and obesity pathogenesis.


CCAAT-Enhancer-Binding Protein-alpha , Carbon , Maillard Reaction , Mesenchymal Stem Cells , PPAR gamma , Sterol Regulatory Element Binding Protein 1 , Humans , Carbon/chemistry , PPAR gamma/genetics , PPAR gamma/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , Quantum Dots/chemistry , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Sulfur/chemistry
2.
Molecules ; 29(9)2024 Apr 28.
Article En | MEDLINE | ID: mdl-38731523

This study reports an innovative approach for producing nanoplastics (NP) from various types of domestic waste plastics without the use of chemicals. The plastic materials used included water bottles, styrofoam plates, milk bottles, centrifuge tubes, to-go food boxes, and plastic bags, comprising polyethylene terephthalate (PET), polystyrene (PS), polypropylene (PP), high-density polyethylene (HDPE), and Poly (Ethylene-co-Methacrylic Acid) (PEMA). The chemical composition of these plastics was confirmed using Raman and FTIR spectroscopy, and they were found to have irregular shapes. The resulting NP particles ranged from 50 to 400 nm in size and demonstrated relative stability when suspended in water. To assess their impact, the study investigated the effects of these NP particulates on cell viability and the expression of genes involved in inflammation and oxidative stress using a macrophage cell line. The findings revealed that all types of NP reduced cell viability in a concentration-dependent manner. Notably, PS, HDPE, and PP induced significant reductions in cell viability at lower concentrations, compared to PEMA and PET. Moreover, exposure to NP led to differential alterations in the expression of inflammatory genes in the macrophage cell line. Overall, this study presents a viable method for producing NP from waste materials that closely resemble real-world NP. Furthermore, the toxicity studies demonstrated distinct cellular responses based on the composition of the NP, shedding light on the potential environmental and health impacts of these particles.


Cell Survival , Macrophages , Microplastics , Cell Survival/drug effects , Macrophages/drug effects , Macrophages/metabolism , Animals , Mice , Nanoparticles/chemistry , Plastics/chemistry , RAW 264.7 Cells , Gene Expression/drug effects , Cell Line , Gene Expression Regulation/drug effects , Waste Products/analysis , Particle Size
3.
Molecules ; 29(9)2024 May 02.
Article En | MEDLINE | ID: mdl-38731597

Fibrosis is a ubiquitous pathology, and prior studies have indicated that various artemisinin (ART) derivatives (including artesunate (AS), artemether (AM), and dihydroartemisinin (DHA)) can reduce fibrosis in vitro and in vivo. The medicinal plant Artemisia annua L. is the natural source of ART and is widely used, especially in underdeveloped countries, to treat a variety of diseases including malaria. A. afra contains no ART but is also antimalarial. Using human dermal fibroblasts (CRL-2097), we compared the effects of A. annua and A. afra tea infusions, ART, AS, AM, DHA, and a liver metabolite of ART, deoxyART (dART), on fibroblast viability and expression of key fibrotic marker genes after 1 and 4 days of treatment. AS, DHA, and Artemisia teas reduced fibroblast viability 4 d post-treatment in up to 80% of their respective controls. After 4 d of treatment, AS DHA and Artemisia teas downregulated ACTA2 up to 10 fold while ART had no significant effect, and AM increased viability by 10%. MMP1 and MMP3 were upregulated by AS, 17.5 and 32.6 fold, respectively, and by DHA, 8 and 51.8 fold, respectively. ART had no effect, but A. annua and A. afra teas increased MMP3 5 and 16-fold, respectively. Although A. afra tea increased COL3A1 5 fold, MMP1 decreased >7 fold with no change in either transcript by A. annua tea. Although A. annua contains ART, it had a significantly greater anti-fibrotic effect than ART alone but was less effective than A. afra. Immunofluorescent staining for smooth-muscle α-actin (α-SMA) correlated well with the transcriptional responses of drug-treated fibroblasts. Together, proliferation, qPCR, and immunofluorescence results show that treatment with ART, AS, DHA, and the two Artemisia teas yield differing responses, including those related to fibrosis, in human dermal fibroblasts, with evidence also of remodeling of fibrotic ECM.


Artemisia , Artemisinins , Fibroblasts , Fibrosis , Humans , Artemisinins/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Artemisia/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Survival/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/genetics , Actins/metabolism , Actins/genetics , Artesunate/pharmacology , Gene Expression Regulation/drug effects , Artemether/pharmacology , Skin/drug effects , Skin/metabolism , Skin/pathology
4.
Anim Biotechnol ; 35(1): 2344208, 2024 Nov.
Article En | MEDLINE | ID: mdl-38741260

Garlic, known for its immune-modulating and antibiotic properties, contains lectins that possess antimicrobial and immunomodulatory effects. Galectins (Gals), which bind ß-galactosides, play a role in modulating immunity and pathological processes. It is hypothesized that garlic's lectin components interfere with animal lectins. St. Croix sheep, known for their resistance to parasites and adaptability, are influenced by dietary supplements for innate immunity. This study evaluated the impact of garlic drench on Galectin gene expression in St. Croix sheep. Adult non-lactating ewes received either garlic juice concentrate or sterile distilled water for four weeks. Blood samples were collected, and plasma and whole blood cells were separated. Galectin secretion was assessed using a Sheep-specific ELISA, while Galectin gene transcription was analyzed through real-time PCR. Garlic administration upregulated LGALS-3 gene expression and significantly increased total plasma protein concentration. Garlic supplementation also affected Galectin secretion, with Gal-1, Gal-3, and Gal-9 showing differential effects.


Galectins , Garlic , Animals , Garlic/chemistry , Galectins/genetics , Galectins/metabolism , Sheep , Female , Dietary Supplements , Gene Expression Regulation/drug effects , Gene Expression/drug effects , Animal Feed/analysis
5.
Pak J Pharm Sci ; 37(1): 79-84, 2024 Jan.
Article En | MEDLINE | ID: mdl-38741403

Vanadyl sulfate (VS), is a component of some food supplements and experimental drugs. This study was carried out to present a novel method for induction of Type 2 diabetes in rats, then for the first time in literature, for evaluating the effect of VS on metabolic parameters and gene expression, simultaneously. 40 male wistar rats were distributed between the four groups, equally. High fat diet and fructose were used for diabetes induction. Diabetic rats treated by two different dose of VS for 12 weeks. Metabolic profiles were evaluated by commercial available kits and gene expression were assayed by real time-PCR. Compared to controls, in non-treated diabetic rats, weight, glucose, triglyceride, total cholesterol, insulin and insulin resistance were increased significantly (p-value <0.05) that indicated induction of type 2 diabetes. Further, the results showed that VS significantly reduced weight, insulin secretion, Tumor Necrosis Factor-alpha (TNF-α) genes expression, lipid profiles except HDL that we couldn't find any significant change and increased Peroxisome Proliferator-Activated Receptor- gamma (PPAR-γ) gene expression in VS-treated diabetic animals in comparison with the non-treated diabetics. Our study demonstrated that vanadyl supplementation in diabetic rats had advantageous effects on metabolic profiles and related gene expression.


Blood Glucose , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , PPAR gamma , Rats, Wistar , Tumor Necrosis Factor-alpha , Vanadium Compounds , Animals , Male , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Blood Glucose/drug effects , Blood Glucose/metabolism , Vanadium Compounds/pharmacology , Insulin Resistance , Rats , Insulin/blood , Hypoglycemic Agents/pharmacology , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects
6.
Biol Open ; 13(5)2024 May 15.
Article En | MEDLINE | ID: mdl-38742438

Bone is increasingly recognized as a target for diabetic complications. In order to evaluate the direct effects of high glucose on bone, we investigated the global transcriptional changes induced by hyperglycemia in osteoblasts in vitro. Rat bone marrow-derived mesenchymal stromal cells were differentiated into osteoblasts for 10 days, and prior to analysis, they were exposed to hyperglycemia (25 mM) for the short-term (1 or 3 days) or long-term (10 days). Genes and pathways regulated by hyperglycemia were identified using mRNA sequencing and verified with qPCR. Genes upregulated by 1-day hyperglycemia were, for example, related to extracellular matrix organization, collagen synthesis and bone formation. This stimulatory effect was attenuated by 3 days. Long-term exposure impaired osteoblast viability, and downregulated, for example, extracellular matrix organization and lysosomal pathways, and increased intracellular oxidative stress. Interestingly, transcriptional changes by different exposure times were mostly unique and only 89 common genes responding to glucose were identified. In conclusion, short-term hyperglycemia had a stimulatory effect on osteoblasts and bone formation, whereas long-term hyperglycemia had a negative effect on intracellular redox balance, osteoblast viability and function.


Gene Expression Regulation , Glucose , Osteoblasts , Osteoblasts/metabolism , Osteoblasts/drug effects , Animals , Glucose/metabolism , Rats , Gene Expression Regulation/drug effects , Gene Expression Profiling , Hyperglycemia/metabolism , Hyperglycemia/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Transcriptome , Osteogenesis/drug effects , Osteogenesis/genetics , Cell Survival/drug effects , Transcription, Genetic/drug effects , Cells, Cultured , Oxidative Stress/drug effects
7.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article En | MEDLINE | ID: mdl-38732159

The receptor for advanced glycation end-products (RAGE) has a central function in orchestrating inflammatory responses in multiple disease states including chronic obstructive pulmonary disease (COPD). RAGE is a transmembrane pattern recognition receptor with particular interest in lung disease due to its naturally abundant pulmonary expression. Our previous research demonstrated an inflammatory role for RAGE following acute exposure to secondhand smoke (SHS). However, chronic inflammatory mechanisms associated with RAGE remain ambiguous. In this study, we assessed transcriptional outcomes in mice exposed to chronic SHS in the context of RAGE expression. RAGE knockout (RKO) and wild-type (WT) mice were delivered nose-only SHS via an exposure system for six months and compared to control mice exposed to room air (RA). We specifically compared WT + RA, WT + SHS, RKO + RA, and RKO + SHS. Analysis of gene expression data from WT + RA vs. WT + SHS showed FEZ1, Slpi, and Msln as significant at the three-month time point; while RKO + SHS vs. WT + SHS identified cytochrome p450 1a1 and Slc26a4 as significant at multiple time points; and the RKO + SHS vs. WT + RA revealed Tmem151A as significant at the three-month time point as well as Gprc5a and Dynlt1b as significant at the three- and six-month time points. Notable gene clusters were functionally analyzed and discovered to be specific to cytoskeletal elements, inflammatory signaling, lipogenesis, and ciliogenesis. We found gene ontologies (GO) demonstrated significant biological pathways differentially impacted by the presence of RAGE. We also observed evidence that the PI3K-Akt and NF-κB signaling pathways were significantly enriched in DEGs across multiple comparisons. These data collectively identify several opportunities to further dissect RAGE signaling in the context of SHS exposure and foreshadow possible therapeutic modalities.


Lung , Mice, Knockout , Receptor for Advanced Glycation End Products , Tobacco Smoke Pollution , Transcriptome , Animals , Receptor for Advanced Glycation End Products/metabolism , Receptor for Advanced Glycation End Products/genetics , Mice , Lung/metabolism , Lung/pathology , Lung/drug effects , Tobacco Smoke Pollution/adverse effects , Mice, Inbred C57BL , Signal Transduction/drug effects , Gene Expression Regulation/drug effects , Male , Gene Expression Profiling
8.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article En | MEDLINE | ID: mdl-38731911

In drug discovery, selecting targeted molecules is crucial as the target could directly affect drug efficacy and the treatment outcomes. As a member of the CCN family, CTGF (also known as CCN2) is an essential regulator in the progression of various diseases, including fibrosis, cancer, neurological disorders, and eye diseases. Understanding the regulatory mechanisms of CTGF in different diseases may contribute to the discovery of novel drug candidates. Summarizing the CTGF-targeting and -inhibitory drugs is also beneficial for the analysis of the efficacy, applications, and limitations of these drugs in different disease models. Therefore, we reviewed the CTGF structure, the regulatory mechanisms in various diseases, and drug development in order to provide more references for future drug discovery.


Connective Tissue Growth Factor , Drug Discovery , Humans , Connective Tissue Growth Factor/metabolism , Drug Discovery/methods , Animals , Neoplasms/drug therapy , Neoplasms/metabolism , Eye Diseases/drug therapy , Eye Diseases/metabolism , Fibrosis , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Gene Expression Regulation/drug effects
9.
Front Immunol ; 15: 1397432, 2024.
Article En | MEDLINE | ID: mdl-38751427

Introduction: The release of mature interleukin (IL-) 1ß from osteoblasts in response to danger signals is tightly regulated by the nucleotide-binding oligomerization domain leucine-rich repeat and pyrin-containing protein 3 (NLRP3) inflammasome. These danger signals include wear products resulting from aseptic loosening of joint arthroplasty. However, inflammasome activation requires two different signals: a nuclear factor-kappa B (NF-κB)-activating priming signal and an actual inflammasome-activating signal. Since human osteoblasts react to wear particles via Toll-like receptors (TLR), particles may represent an inflammasome activator that can induce both signals. Methods: Temporal gene expression profiles of TLRs and associated intracellular signaling pathways were determined to investigate the period when human osteoblasts take up metallic wear particles after initial contact and initiate a molecular response. For this purpose, human osteoblasts were treated with metallic particles derived from cobalt-chromium alloy (CoCr), lipopolysaccharides (LPS), and tumor necrosis factor-alpha (TNF) alone or in combination for incubation times ranging from one hour to three days. Shortly after adding the particles, their uptake was observed by the change in cell morphology and spectral data. Results: Exposure of osteoblasts to particles alone increased NLRP3 inflammasome-associated genes. The response was not significantly enhanced when cells were treated with CoCr + LPS or CoCr + TNF, whereas inflammation markers were induced. Despite an increase in genes related to the NLRP3 inflammasome, the release of IL-1ß was unaffected after contact with CoCr particles. Discussion: Although CoCr particles affect the expression of NLRP3 inflammasome-associated genes, a single stimulus was not sufficient to prime and activate the inflammasome. TNF was able to prime the NLRP3 inflammasome of human osteoblasts.


Gene Expression Regulation , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Osteoblasts , Tumor Necrosis Factor-alpha , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteoblasts/immunology , Inflammasomes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Gene Expression Regulation/drug effects , Cells, Cultured , Signal Transduction/drug effects
10.
Aquat Toxicol ; 271: 106925, 2024 Jun.
Article En | MEDLINE | ID: mdl-38718521

Excessive antibiotic use has led to the spread of antibiotic resistance genes (ARGs), impacting gut microbiota and host health. However, the effects of antibiotics on amphibian populations remain unclear. We investigated the impact of oxytetracycline (OTC) and ciprofloxacin (CIP) on Chinese giant salamanders (Andrias davidianus), focusing on gut microbiota, ARGs, and gene expression by performing metagenome and transcriptome sequencing. A. davidianus were given OTC (20 or 40 mg/kg) or CIP (50 or 100 mg/kg) orally for 7 days. The results revealed that oral administration of OTC and CIP led to distinct changes in microbial composition and functional potential, with CIP treatment having a greater impact than OTC. Antibiotic treatment also influenced the abundance of ARGs, with an increase in fluoroquinolone and multi-drug resistance genes observed post-treatment. The construction of metagenome-assembled genomes (MAGs) accurately validated that CIP intervention enriched fish-associated potential pathogens Aeromonas hydrophila carrying an increased number of ARGs. Additionally, mobile genetic elements (MGEs), such as phages and plasmids, were implicated in the dissemination of ARGs. Transcriptomic analysis of the gut revealed significant alterations in gene expression, particularly in immune-related pathways, with differential effects observed between OTC and CIP treatments. Integration of metagenomic and transcriptomic data highlighted potential correlations between gut gene expression and microbial composition, suggesting complex interactions between the host gut and its gut microbiota in response to antibiotic exposure. These findings underscore the importance of understanding the impact of antibiotic intervention on the gut microbiome and host health in amphibians, particularly in the context of antibiotic resistance and immune function.


Anti-Bacterial Agents , Ciprofloxacin , Gastrointestinal Microbiome , Oxytetracycline , Urodela , Animals , Oxytetracycline/toxicity , Gastrointestinal Microbiome/drug effects , Ciprofloxacin/pharmacology , Ciprofloxacin/toxicity , Urodela/genetics , Urodela/microbiology , Anti-Bacterial Agents/toxicity , Anti-Bacterial Agents/pharmacology , Transcriptome/drug effects , Metagenome , Metagenomics , Gene Expression Profiling , Water Pollutants, Chemical/toxicity , Aeromonas hydrophila/drug effects , Gene Expression Regulation/drug effects
11.
Sci Rep ; 14(1): 10733, 2024 05 10.
Article En | MEDLINE | ID: mdl-38730024

Molecular responses to alcohol consumption are dynamic, context-dependent, and arise from a complex interplay of biological and external factors. While many have studied genetic risk associated with drinking patterns, comprehensive studies identifying dynamic responses to pharmacologic and psychological/placebo effects underlying binge drinking are lacking. We investigated transcriptome-wide response to binge, medium, and placebo alcohol consumption by 17 healthy heavy social drinkers enrolled in a controlled, in-house, longitudinal study of up to 12 days. Using RNA-seq, we identified 251 and 13 differentially expressed genes (DEGs) in response to binge drinking and placebo, respectively. Eleven protein-coding DEGs had very large effect sizes in response to binge drinking (Cohen's d > 1). Furthermore, binge dose significantly impacted the Cytokine-cytokine receptor interaction pathway (KEGG: hsa04060) across all experimental sequences. Placebo also impacted hsa04060, but only when administered following regular alcohol drinking sessions. Similarly, medium-dose and placebo commonly impacted KEGG pathways of Systemic lupus erythematosus, Neutrophil extracellular trap formation, and Alcoholism based on the sequence of drinking sessions. These findings together indicate the "dose-extending effects" of placebo at a molecular level. Furthermore, besides supporting alcohol dose-specific molecular changes, results suggest that the placebo effects may induce molecular responses within the same pathways regulated by alcohol.


Binge Drinking , Gene Expression Profiling , Placebo Effect , Transcriptome , Humans , Binge Drinking/blood , Binge Drinking/genetics , Male , Female , Adult , Young Adult , Ethanol , Longitudinal Studies , Gene Expression Regulation/drug effects
12.
Life Sci Alliance ; 7(7)2024 Jul.
Article En | MEDLINE | ID: mdl-38697845

Defective mitophagy in renal tubular epithelial cells is one of the main drivers of renal fibrosis in diabetic kidney disease. Our gene sequencing data showed the expression of PINK1 and BNIP3, two key molecules of mitophagy, was decreased in renal tissues of VDR-knockout mice. Herein, streptozotocin (STZ) was used to induce renal interstitial fibrosis in mice. VDR deficiency exacerbated STZ-induced renal impairment and defective mitophagy. Paricalcitol (pari, a VDR agonist) and the tubular epithelial cell-specific overexpression of VDR restored the expression of PINK1 and BNIP3 in the renal cortex and attenuated STZ-induced kidney fibrosis and mitochondrial dysfunction. In HK-2 cells under high glucose conditions, an increased level of α-SMA, COL1, and FN and a decreased expression of PINK1 and BNIP3 with severe mitochondrial damage were observed, and these alterations could be largely reversed by pari treatment. ChIP-qPCR and luciferase reporter assays showed VDR could positively regulate the transcription of Pink1 and Bnip3 genes. These findings reveal that VDR could restore mitophagy defects and attenuate STZ-induced fibrosis in diabetic mice through regulation of PINK1 and BNIP3.


Diabetes Mellitus, Experimental , Diabetic Nephropathies , Ergocalciferols , Membrane Proteins , Mice, Knockout , Mitophagy , Protein Kinases , Receptors, Calcitriol , Streptozocin , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , Mitophagy/genetics , Mitophagy/drug effects , Protein Kinases/metabolism , Protein Kinases/genetics , Humans , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/genetics , Male , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Fibrosis , Kidney Tubules/metabolism , Kidney Tubules/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Mice, Inbred C57BL , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Cell Line , Gene Expression Regulation/drug effects
13.
Neuromolecular Med ; 26(1): 18, 2024 May 01.
Article En | MEDLINE | ID: mdl-38691185

Seipin is a key regulator of lipid metabolism, the deficiency of which leads to severe lipodystrophy. Hypothalamus is the pivotal center of brain that modulates appetite and energy homeostasis, where Seipin is abundantly expressed. Whether and how Seipin deficiency leads to systemic metabolic disorders via hypothalamus-involved energy metabolism dysregulation remains to be elucidated. In the present study, we demonstrated that Seipin-deficiency induced hypothalamic inflammation, reduction of anorexigenic pro-opiomelanocortin (POMC), and elevation of orexigenic agonist-related peptide (AgRP). Importantly, administration of rosiglitazone, a thiazolidinedione antidiabetic agent, rescued POMC and AgRP expression, suppressed hypothalamic inflammation, and restored energy homeostasis in Seipin knockout mice. Our findings offer crucial insights into the mechanism of Seipin deficiency-associated energy imbalance and indicates that rosiglitazone could serve as potential intervening agent towards metabolic disorders linked to Seipin.


Agouti-Related Protein , Energy Metabolism , GTP-Binding Protein gamma Subunits , Homeostasis , Hypothalamus , Mice, Knockout , Pro-Opiomelanocortin , Rosiglitazone , Animals , Mice , Hypothalamus/metabolism , Energy Metabolism/drug effects , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/biosynthesis , Agouti-Related Protein/genetics , GTP-Binding Protein gamma Subunits/genetics , Rosiglitazone/pharmacology , Male , Neuroinflammatory Diseases/etiology , Mice, Inbred C57BL , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Neuropeptides/genetics , Neuropeptides/deficiency , Gene Expression Regulation/drug effects
14.
Life Sci Alliance ; 7(8)2024 Aug.
Article En | MEDLINE | ID: mdl-38755006

Diabetes complications such as nephropathy, retinopathy, or cardiovascular disease arise from vascular dysfunction. In this context, it has been observed that past hyperglycemic events can induce long-lasting alterations, a phenomenon termed "metabolic memory." In this study, we evaluated the genome-wide gene expression and chromatin accessibility alterations caused by transient high-glucose exposure in human endothelial cells (ECs) in vitro. We found that cells exposed to high glucose exhibited substantial gene expression changes in pathways known to be impaired in diabetes, many of which persist after glucose normalization. Chromatin accessibility analysis also revealed that transient hyperglycemia induces persistent alterations, mainly in non-promoter regions identified as enhancers with neighboring genes showing lasting alterations. Notably, activation of the NRF2 pathway through NRF2 overexpression or supplementation with the plant-derived compound sulforaphane, effectively reverses the glucose-induced transcriptional and chromatin accessibility memories in ECs. These findings underscore the enduring impact of transient hyperglycemia on ECs' transcriptomic and chromatin accessibility profiles, emphasizing the potential utility of pharmacological NRF2 pathway activation in mitigating and reversing the high-glucose-induced transcriptional and epigenetic alterations.


Epigenesis, Genetic , Glucose , NF-E2-Related Factor 2 , Signal Transduction , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Humans , Glucose/metabolism , Epigenesis, Genetic/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Hyperglycemia/metabolism , Hyperglycemia/genetics , Chromatin/metabolism , Chromatin/genetics , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Transcription, Genetic/drug effects , Gene Expression Regulation/drug effects , Isothiocyanates/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Sulfoxides/pharmacology
15.
PLoS One ; 19(5): e0303528, 2024.
Article En | MEDLINE | ID: mdl-38753618

Arsenic has been identified as an environmental toxicant acting through various mechanisms, including the disruption of endocrine pathways. The present study assessed the ability of a single intraperitoneal injection of arsenic, to modify the mRNA expression levels of estrogen- and thyroid hormone receptors (ERα,ß; TRα,ß) and peroxisome proliferator-activated receptor gamma (PPARγ) in hypothalamic tissue homogenates of prepubertal mice in vivo. Mitochondrial respiration (MRR) was also measured, and the corresponding mitochondrial ultrastructure was analyzed. Results show that ERα,ß, and TRα expression was significantly increased by arsenic, in all concentrations examined. In contrast, TRß and PPARγ remained unaffected after arsenic injection. Arsenic-induced dose-dependent changes in state 4 mitochondrial respiration (St4). Mitochondrial morphology was affected by arsenic in that the 5 mg dose increased the size but decreased the number of mitochondria in agouti-related protein- (AgRP), while increasing the size without affecting the number of mitochondria in pro-opiomelanocortin (POMC) neurons. Arsenic also increased the size of the mitochondrial matrix per host mitochondrion. Complex analysis of dose-dependent response patterns between receptor mRNA, mitochondrial morphology, and mitochondrial respiration in the neuroendocrine hypothalamus suggests that instant arsenic effects on receptor mRNAs may not be directly reflected in St3-4 values, however, mitochondrial dynamics is affected, which predicts more pronounced effects in hypothalamus-regulated homeostatic processes after long-term arsenic exposure.


Arsenic , Hypothalamus , Mitochondria , PPAR gamma , RNA, Messenger , Animals , Hypothalamus/metabolism , Hypothalamus/drug effects , Mice , Mitochondria/metabolism , Mitochondria/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics , Arsenic/toxicity , Receptors, Thyroid Hormone/metabolism , Receptors, Thyroid Hormone/genetics , Male , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Cell Respiration/drug effects , Gene Expression Regulation/drug effects
16.
Mol Neurodegener ; 19(1): 37, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38654375

BACKGROUND: Microglia play important roles in maintaining brain homeostasis and neurodegeneration. The discovery of genetic variants in genes predominately or exclusively expressed in myeloid cells, such as Apolipoprotein E (APOE) and triggering receptor expressed on myeloid cells 2 (TREM2), as the strongest risk factors for Alzheimer's disease (AD) highlights the importance of microglial biology in the brain. The sequence, structure and function of several microglial proteins are poorly conserved across species, which has hampered the development of strategies aiming to modulate the expression of specific microglial genes. One way to target APOE and TREM2 is to modulate their expression using antisense oligonucleotides (ASOs). METHODS: In this study, we identified, produced, and tested novel, selective and potent ASOs for human APOE and TREM2. We used a combination of in vitro iPSC-microglia models, as well as microglial xenotransplanted mice to provide proof of activity in human microglial in vivo. RESULTS: We proved their efficacy in human iPSC microglia in vitro, as well as their pharmacological activity in vivo in a xenografted microglia model. We demonstrate ASOs targeting human microglia can modify their transcriptional profile and their response to amyloid-ß plaques in vivo in a model of AD. CONCLUSIONS: This study is the first proof-of-concept that human microglial can be modulated using ASOs in a dose-dependent manner to manipulate microglia phenotypes and response to neurodegeneration in vivo.


Alzheimer Disease , Microglia , Oligonucleotides, Antisense , Microglia/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Humans , Oligonucleotides, Antisense/pharmacology , Animals , Mice , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Induced Pluripotent Stem Cells/metabolism , Gene Expression Regulation/drug effects , Disease Models, Animal
17.
PLoS One ; 19(4): e0301904, 2024.
Article En | MEDLINE | ID: mdl-38662666

Our previous research demonstrated that PU.1 regulates expression of the genes involved in inflammation in macrophages. Selective knockdown of PU.1 in macrophages ameliorated LPS-induced acute lung injury (ALI) in bone marrow chimera mice. Inhibitors that block the transcriptional activity of PU.1 in macrophages have the potential to mitigate the pathophysiology of LPS-induced ALI. However, complete inactivation of PU.1 gene disrupts normal myelopoiesis. Although the green tea polyphenol Epigallocatechin gallate (EGCG) has been shown to regulate inflammatory genes in various cell types, it is not known if EGCG alters the transcriptional activity of PU.1 protein. Using Schrodinger Glide docking, we have identified that EGCG binds with PU.1 protein, altering its DNA-binding and self-dimerization activity. In silico analysis shows that EGCG forms Hydrogen bonds with Glutamic Acid 209, Leucine 250 in DNA binding and Lysine 196, Tryptophan 193, and Leucine 182 in the self-dimerization domain of the PU.1 protein. Experimental validation using mouse bone marrow-derived macrophages (BMDM) confirmed that EGCG inhibits both DNA binding by PU.1 and self-dimerization. Importantly, EGCG had no impact on expression of the total PU.1 protein levels but significantly reduced expression of various inflammatory genes and generation of ROS. In summary, we report that EGCG acts as an inhibitor of the PU.1 transcription factor in macrophages.


Catechin , Catechin/analogs & derivatives , Macrophages , Proto-Oncogene Proteins , Trans-Activators , Catechin/pharmacology , Animals , Trans-Activators/metabolism , Trans-Activators/genetics , Macrophages/metabolism , Macrophages/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Protein Binding , DNA/metabolism , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology
18.
Article En | MEDLINE | ID: mdl-38663833

Disruption of the thyroid hormone system by synthetic chemicals is gaining attention owing to its potential negative effects on organisms. In this study, the effects of the dio-inhibitor iopanoic acid (IOP) on the levels of thyroid hormone and related gene expression, swim bladder inflation, and swimming performance were investigated in Japanese medaka. Iopanoic acid exposure suppressed thyroid-stimulating hormone ß (tshß), tshß-like, iodotyronin deiodinase 1 (dio1), and dio2 expression, and increased T4 and T3 levels. In addition, IOP exposure inhibited swim bladder inflation, reducing swimming performance. Although adverse outcome pathways of thyroid hormone disruption have been developed using zebrafish, no adverse outcome pathways have been developed using Japanese medaka. This study confirmed that IOP inhibits dio expression (a molecular initiating event), affects T3 and T4 levels (a key event), and reduces swim bladder inflation (a key event) and swimming performance (an adverse outcome) in Japanese medaka.


Air Sacs , Iopanoic Acid , Oryzias , Swimming , Thyroid Hormones , Animals , Oryzias/physiology , Air Sacs/drug effects , Air Sacs/metabolism , Thyroid Hormones/metabolism , Thyroid Hormones/blood , Iopanoic Acid/toxicity , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression Regulation/drug effects , Thyroxine/blood , Triiodothyronine/blood , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism
19.
Gene ; 916: 148446, 2024 Jul 20.
Article En | MEDLINE | ID: mdl-38583816

Mesenchymal stem cells (MSCs) have high priority in clinical applications for treatment of immune disorders because of their immunomodulatory function. A lot of researches have currently been undertaken to enhance the stemness capacities of the cells and pick an excellent type of MSCs for clinical approaches. This study aims to assess the immunomodulatory related MicroRNAs (miRNAs) expression as well as their target genes in both adipose derived stem cells (Ad-SCs) and dental pulp derived stem cell (DP-SCs) in the presence or lack of Crocin (saffron plant's bioactive compound). For this purpose, first MSCs were extracted from adipose and dental pulp tissues, and then their mesenchymal nature was confirmed using flow cytometry and differentiation tests. Following the cell treatment with an optimal-non-toxic dose of Crocin (Obtained by MTT test), the expression of 4 selected immunomodulatory-related micro-RNAs (Mir-126, -21, -23, and-155) and their target genes (PI3K/ Akt 1 and 2/ NFKB and RELA) were assessed by RT-PCR. Our findings revealed that miRNA-23 and miRNA-126 were up-regulated in both types of cells treated with Crocin, while in the other side, miRNA-21 and miRNA-155 were down-regulated in DP-SCs and were up-regulated in Ad-SCs under treatment. Moreover, the real-time PCR results indicated that Crocin could significantly down regulate the expression of PI3K/ Akt1/ Akt2/ NFKB/ RELA genes in DP-SCs and PI3K/Akt2 genes in Ad-SCs and up regulate the expression of Akt1/ NFKB/ RELA genes in recent cells. Based on the analysis of the obtained data, the immunoregulatory effects of Crocin were higher in DP-SCs than in Ad-SCs. In conclusion, Crocin could control essential signaling pathways related to the inflammation by regulating the expression of related- miRNAs genes that play a key function in the immune regulation pathways in MSCs. Our findings can give an understanding of the mechanisms by which Crocin enhances the immunomodulatory feature of MSCs. According to the research findings, DP-SCs are probably a better immunomodulator in Crocin treatment than Ad-SCs and it may be helpful for MSCs selection in clinical applications for modulation or treatment of autoimmune disorders.


Carotenoids , Mesenchymal Stem Cells , MicroRNAs , MicroRNAs/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Carotenoids/pharmacology , Humans , Cells, Cultured , Gene Expression Regulation/drug effects , Cell Differentiation/drug effects , Immunomodulation/drug effects , Immunomodulation/genetics , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Adipose Tissue/cytology , Adipose Tissue/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism
20.
Free Radic Biol Med ; 219: 153-162, 2024 Jul.
Article En | MEDLINE | ID: mdl-38657753

The anemia of inflammation (AI) is characterized by the presence of inflammation and abnormal elevation of hepcidin. Accumulating evidence has proved that Rocaglamide (RocA) was involved in inflammation regulation. Nevertheless, the role of RocA in AI, especially in iron metabolism, has not been investigated, and its underlying mechanism remains elusive. Here, we demonstrated that RocA dramatically suppressed the elevation of hepcidin and ferritin in LPS-treated mice cell line RAW264.7 and peritoneal macrophages. In vivo study showed that RocA can restrain the depletion of serum iron (SI) and transferrin (Tf) saturation caused by LPS. Further investigation showed that RocA suppressed the upregulation of hepcidin mRNA and downregulation of Fpn1 protein expression in the spleen and liver of LPS-treated mice. Mechanistically, this effect was attributed to RocA's ability to inhibit the IL-6/STAT3 pathway, resulting in the suppression of hepcidin mRNA and subsequent increase in Fpn1 and TfR1 expression in LPS-treated macrophages. Moreover, RocA inhibited the elevation of the cellular labile iron pool (LIP) and reactive oxygen species (ROS) induced by LPS in RAW264.7 cells. These findings reveal a pivotal mechanism underlying the roles of RocA in modulating iron homeostasis and also provide a candidate natural product on alleviating AI.


Hepcidins , Homeostasis , Interleukin-6 , Iron , Lipopolysaccharides , Receptors, Transferrin , STAT3 Transcription Factor , Hepcidins/metabolism , Hepcidins/genetics , Animals , Mice , Iron/metabolism , RAW 264.7 Cells , Receptors, Transferrin/metabolism , Receptors, Transferrin/genetics , Lipopolysaccharides/pharmacology , Interleukin-6/metabolism , Interleukin-6/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Reactive Oxygen Species/metabolism , Gene Expression Regulation/drug effects , Inflammation/metabolism , Inflammation/genetics , Inflammation/pathology , Signal Transduction/drug effects , Anemia/metabolism , Anemia/genetics , Anemia/drug therapy , Anemia/pathology , Ferritins/metabolism , Ferritins/genetics , Male , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Macrophages/drug effects , Cation Transport Proteins
...