Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 375
Filter
1.
PLoS One ; 19(8): e0309166, 2024.
Article in English | MEDLINE | ID: mdl-39172905

ABSTRACT

Chronic obstructive pulmonary disease is a common chronic lung disease with an ever-increasing incidence. Despite years of drug research and approvals, we are still not able to halt progress or restore normal lung function. Our previous studies have demonstrated that liver growth factor-LGF has an effect on the repair of the affected tissue in a mouse model of cigarette smoke exposure, but by what pathways it achieves this is unknown. The present study aimed to identify differentially expressed genes between emphysematous mice treated with LGF to identify potential therapeutic targets for the treatment of pulmonary emphysema. The emphysema mouse model was induced by prolonged exposure to cigarette smoke. To determine the gene expression profile of the lung in smokers treated or not with LGF, lung messenger RNA gene expression was assessed with the Agilent Array platform. We carried out differentially expressed gene analysis, functional enrichment and validated in treated mouse lung samples. The treated group significantly improved lung function (~35%) and emphysema level (~20%), consistent with our previous published studies. Microarray analysis demonstrated 290 differentially expressed genes in total (2.0-fold over or lower expressed). Injury repair-associated genes and pathways were further enhanced in the lung of LGF treated mice. The expression trends of two genes (Zscan2 and Bag6) were different in emphysematous lungs treated with LGF compared to untreated lungs. Therefore, Zscan2 and Bag6 genes could play a role in regulating inflammation and the immune response in the lung that undergoes partial lung regeneration. However, further studies are necessary to demonstrate this causal relationship.


Subject(s)
Disease Models, Animal , Lung , Pulmonary Disease, Chronic Obstructive , Transcription Factors , Animals , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Mice , Lung/metabolism , Lung/drug effects , Lung/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Gene Expression Profiling , Mice, Inbred C57BL , Male , Gene Expression Regulation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
2.
Matrix Biol ; 132: 47-58, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39147560

ABSTRACT

BACKGROUND: Lung fibroblasts play a central role in maintaining lung homeostasis and facilitating repair through the synthesis and organization of the extracellular matrix (ECM). This study investigated the cross-talk between interleukin-1 alpha (IL-1α) and transforming growth factor-ß (TGF-ß) signaling, two key regulators in tissue repair and fibrosis, in the context of lung fibroblast repair in the healthy lung. RESULTS: Stimulation of lung fibroblasts with TGF-ß1 and TGF-ß2 induced collagen-I and fibronectin protein expression (p < 0.05), a response inhibited with co-treatment with IL-1α (p < 0.05). Additionally, TGF-ß1 and TGF-ß2 induced myofibroblast differentiation, and collagen-I gel contraction, which were both suppressed by IL-1α (p < 0.05). In contrast, interleukin (IL)-6, IL-8 and thymic stromal lymphopoietin induced by IL-1α, were unaffected by TGF-ß1 or TGF-ß2. Mechanistically, IL-1α administration led to the suppression of TGF-ß1 and TGF-ß2 signaling, through downregulation of mRNA and protein for TGF-ß receptor II and the downstream adaptor protein TRAF6, but not through miR-146a that is known to be induced by IL-1α. DISCUSSION: IL-1α acts as a master regulator, modulating TGF-ß1 and TGF-ß2-induced ECM production, remodeling, and myofibroblast differentiation in human lung fibroblasts, playing a vital role in balancing tissue repair versus fibrosis. Further research is required to understand the dysregulated cross-talk between IL-1α and TGF-ß signaling in chronic lung diseases and the exploration of therapeutic opportunities. METHODS: Primary human lung fibroblasts (PHLF) were treated with media control, or 1 ng/ml IL-1α with or without 50 ng/ml TGF-ß1 or TGF-ß2 for 1, 6 and 72 h. Cell lysates were assessed for the expression of ECM proteins and signaling molecules by western blot, miRNA by qPCR, mRNA by RNA sequencing and cell supernatants for cytokine production by ELISA. PHLFs were also seeded in non-tethered collagen-I gels to measure contraction, and myofibroblast differentiation using confocal microscopy.


Subject(s)
Extracellular Matrix , Fibroblasts , Interleukin-1alpha , Lung , Signal Transduction , Transforming Growth Factor beta1 , Humans , Interleukin-1alpha/metabolism , Interleukin-1alpha/genetics , Extracellular Matrix/metabolism , Transforming Growth Factor beta1/metabolism , Lung/metabolism , Lung/cytology , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/cytology , Cell Differentiation , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Cells, Cultured , Collagen Type I/metabolism , Collagen Type I/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Fibronectins/metabolism , Fibronectins/genetics , Gene Expression Regulation/drug effects , Transforming Growth Factor beta2
3.
Sci Rep ; 14(1): 18962, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39152192

ABSTRACT

Cadmium, a common metal pollutant, has been demonstrated to induce type 2 diabetes by disrupting pancreatic ß cells function. In this study, transcriptome microarray was utilized to identify differential gene expression in oxidative damage to pancreatic ß cells following cadmium exposure. The results indicated that a series of mRNAs, LncRNAs, and miRNAs were altered. Of the differentially expressed miRNAs, miR-29a-3p exhibited the most pronounced alteration, with an 11.62-fold increase relative to the control group. Following this, the target gene of miR-29a-3p was identified as Col3a1 through three databases (miRDB, miRTarbase and Tarbase), which demonstrated a decrease across the transcriptome microarray. The upstream target gene of miR-29a-3p was identified as NONMMUT036805, with decreased expression observed in the microarray. Finally, the expression trend of NONMMUT036805/miR-29a-3p/Col3a1 was reversed following NAC pretreatment. This was accompanied by a reduction in oxidative damage indicators, MDA/ROS/GSH-Px appeared to be negatively affected to varying degrees. In conclusion, this study has demonstrated that multiple RNAs are altered during cadmium exposure-induced oxidative damage in pancreatic ß cells. The NONMMUT036805/miR-29a-3p/Col3a1 axis has been shown to be involved in this process, which provides a foundation for the identification of potential targets for cadmium toxicity intervention.


Subject(s)
Cadmium , Insulin-Secreting Cells , MicroRNAs , Oxidative Stress , RNA, Competitive Endogenous , Animals , Mice , Cadmium/toxicity , Cell Line , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , RNA, Competitive Endogenous/genetics , RNA, Competitive Endogenous/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome
4.
J Cell Mol Med ; 28(16): e18588, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39153206

ABSTRACT

Huntington's disease (HD) is a gradually severe neurodegenerative ailment characterised by an increase of a specific trinucleotide repeat sequence (cytosine-adenine-guanine, CAG). It is passed down as a dominant characteristic that worsens over time, creating a significant risk. Despite being monogenetic, the underlying mechanisms as well as biomarkers remain poorly understood. Furthermore, early detection of HD is challenging, and the available diagnostic procedures have low precision and accuracy. The research was conducted to provide knowledge of the biomarkers, pathways and therapeutic targets involved in the molecular processes of HD using informatic based analysis and applying network-based systems biology approaches. The gene expression profile datasets GSE97100 and GSE74201 relevant to HD were studied. As a consequence, 46 differentially expressed genes (DEGs) were identified. 10 hub genes (TPM1, EIF2S3, CCN2, ACTN1, ACTG2, CCN1, CSRP1, EIF1AX, BEX2 and TCEAL5) were further differentiated in the protein-protein interaction (PPI) network. These hub genes were typically down-regulated. Additionally, DEGs-transcription factors (TFs) connections (e.g. GATA2, YY1 and FOXC1), DEG-microRNA (miRNA) interactions (e.g. hsa-miR-124-3p and has-miR-26b-5p) were also comprehensively forecast. Additionally, related gene ontology concepts (e.g. sequence-specific DNA binding and TF activity) connected to DEGs in HD were identified using gene set enrichment analysis (GSEA). Finally, in silico drug design was employed to find candidate drugs for the treatment HD, and while the possible modest therapeutic compounds (e.g. cortistatin A, 13,16-Epoxy-25-hydroxy-17-cheilanthen-19,25-olide, Hecogenin) against HD were expected. Consequently, the results from this study may give researchers useful resources for the experimental validation of Huntington's diagnosis and therapeutic approaches.


Subject(s)
Computational Biology , Gene Regulatory Networks , Huntington Disease , Protein Interaction Maps , Huntington Disease/genetics , Huntington Disease/drug therapy , Huntington Disease/metabolism , Humans , Computational Biology/methods , Protein Interaction Maps/genetics , Protein Interaction Maps/drug effects , Gene Expression Profiling , Biomarkers/metabolism , Gene Expression Regulation/drug effects , Molecular Targeted Therapy , Transcriptome/genetics , Gene Ontology , MicroRNAs/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Mol Med ; 30(1): 125, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152382

ABSTRACT

BACKGROUND: Epimedin A (EA) has been shown to suppress extensive osteoclastogenesis and bone resorption, but the effects of EA remain incompletely understood. The aim of our study was to investigate the effects of EA on osteoclastogenesis and bone resorption to explore the corresponding signalling pathways. METHODS: Rats were randomly assigned to the sham operation or ovariectomy group, and alendronate was used for the positive control group. The therapeutic effect of EA on osteoporosis was systematically analysed by measuring bone mineral density and bone biomechanical properties. In vitro, RAW264.7 cells were treated with receptor activator of nuclear factor kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) to induce osteoclast differentiation. Cell viability assays, tartrate-resistant acid phosphatase (TRAP) staining, and immunofluorescence were used to elucidate the effects of EA on osteoclastogenesis. In addition, the expression of bone differentiation-related proteins or genes was evaluated using Western blot analysis or quantitative polymerase chain reaction (PCR), respectively. RESULTS: After 3 months of oral EA intervention, ovariectomized rats exhibited increased bone density, relative bone volume, trabecular thickness, and trabecular number, as well as reduced trabecular separation. EA dose-dependently normalized bone density and trabecular microarchitecture in the ovariectomized rats. Additionally, EA inhibited the expression of TRAP and NFATc1 in the ovariectomized rats. Moreover, the in vitro results indicated that EA inhibits osteoclast differentiation by suppressing the TRAF6/PI3K/AKT/NF-κB pathway. Further studies revealed that the effect on osteoclast differentiation, which was originally inhibited by EA, was reversed when the TRAF6 gene was overexpressed. CONCLUSIONS: The findings indicated that EA can negatively regulate osteoclastogenesis by inhibiting the TRAF6/PI3K/AKT/NF-κB axis and that ameliorating ovariectomy-induced osteoporosis in rats with EA may be a promising potential therapeutic strategy for the treatment of osteoporosis.


Subject(s)
Cell Differentiation , NF-kappa B , Osteoclasts , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TNF Receptor-Associated Factor 6 , Animals , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , Osteoclasts/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Cell Differentiation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Female , Phosphatidylinositol 3-Kinases/metabolism , Rats , Mice , RAW 264.7 Cells , Flavonoids/pharmacology , Osteogenesis/drug effects , Rats, Sprague-Dawley , Osteoporosis/metabolism , Osteoporosis/etiology , Ovariectomy/adverse effects , Gene Expression Regulation/drug effects , Bone Density/drug effects
6.
J Neuroinflammation ; 21(1): 206, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160534

ABSTRACT

Since its detection in the brain, the cannabinoid receptor type 2 (CB2) has been considered a promising therapeutic target for various neurological and psychiatric disorders. However, precise brain mapping of its expression is still lacking. Using magnetic cell sorting, calibrated RT-qPCR and single-nucleus RNAseq, we show that CB2 is expressed at a low level in all brain regions studied, mainly by few microglial cells, and by neurons in an even lower proportion. Upon lipopolysaccharide stimulation, modeling neuroinflammation in non-sterile conditions, we demonstrate that the inflammatory response is associated with a transient reduction in CB2 mRNA levels in brain tissue, particularly in microglial cells. This result, confirmed in the BV2 microglial cell line, contrasts with the positive correlation observed between CB2 mRNA levels and the inflammatory response upon stimulation by interferon-gamma, modeling neuroinflammation in sterile condition. Discrete brain CB2 expression might thus be up- or down-regulated depending on the inflammatory context.


Subject(s)
Brain , Lipopolysaccharides , Mice, Inbred C57BL , Microglia , Receptor, Cannabinoid, CB2 , Animals , Microglia/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/biosynthesis , Mice , Brain/metabolism , Lipopolysaccharides/pharmacology , Male , Gene Expression Regulation/drug effects , Inflammation/metabolism , Inflammation/pathology , Neuroinflammatory Diseases/metabolism
7.
BMC Vet Res ; 20(1): 344, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097704

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) induces a poor innate immune response following infection. This study evaluates the effects of transforming growth factor beta 1 (TGFß1) up-regulated by PRRSV on gene expressions of co-stimulatory molecules, type I interferon (IFN), type I IFN-regulated genes (IRGs), pattern recognition receptors, and pro-inflammatory cytokines in PRRSV-inoculated monocyte-derived macrophages (MDMs). Phosphorothioate-modified antisense oligodeoxynucleotides (AS ODNs) specific to various regions of porcine TGFß1 mRNA were synthesized, and those specific to the AUG region efficiently knockdown TGFß1 mRNA expression and protein translation. Transfection of TGFßAS ODNs in MDMs inoculated with either classical PRRSV-2 (cPRRSV-2) or highly pathogenic PRRSV-2 (HP-PRRSV-2) significantly reduced TGFß1 mRNA expression and significantly increased mRNA expressions of CD80, CD86, IFNß, IRGs (i.e. IFN regulatory factor 3 (IRF3), IRF7, myxovirus resistance 1, osteopontin, and stimulator of IFN genes), Toll-like receptor 3, and tumor necrosis factor-alpha. Transfection of TGFßAS ODNs in MDMs inoculated with HP-PRRSV-2 also significantly increased mRNA expressions of IFNα, IFNγ, and 2'-5'-oligoadenylate synthetase 1. The quantity of PRRSV-2 RNA copy numbers was significantly reduced in MDMs transfected with TGFßAS ODNs as compared to untransfected MDMs. Recombinant porcine TGFß1 (rTGFß1) and recombinant porcine IFNα (rIFNα) sustained and reduced the yields of PRRSV-2 RNA copy numbers in PRRSV-2 inoculated MDMs, respectively. These findings demonstrate a strategy of PRRSV for innate immune suppression via an induction of TGFß expression. These findings also suggest TGFß as a potential parameter that future PRRSV vaccine and vaccine adjuvant candidates should take into consideration.


Subject(s)
Cytokines , Interferon Type I , Macrophages , Porcine respiratory and reproductive syndrome virus , Animals , Porcine respiratory and reproductive syndrome virus/physiology , Swine , Interferon Type I/metabolism , Cytokines/genetics , Cytokines/metabolism , Macrophages/drug effects , Macrophages/virology , Macrophages/immunology , Macrophages/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Gene Expression Regulation/drug effects , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/virology , Gene Knockdown Techniques , Immunity, Innate
8.
Mol Biol Rep ; 51(1): 909, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39145884

ABSTRACT

BACKGROUND: Inflammatory cytokines such as Interleukin 1ß(IL1ß), IL6,Tumor Necrosis Factor-α (TNF-α) can inhibit osteoblast differentiation and induce osteoblast apoptosis. PANoptosis, a newly identified type of programmed cell death (PCD), may be influenced by long noncoding RNA (lncRNAs) which play important roles in regulating inflammation. However, the potential role of lncRNAs in inflammation and PANoptosis during osteogenic differentiation remains unclear. This study aimed to investigate the regulatory functions of lncRNAs in inflammation and apoptosis during osteogenic differentiation. METHODS AND RESULTS: High-throughput sequencing was used to identify differentially expressed genes involved in osteoblast differentiation under inflammatory conditions. Two lncRNAs associated with inflammation and PANoptosis during osteogenic differentiation were identified from sequencing data and Gene Expression Omnibus (GEO) databases. Their functionalities were analyzed using diverse bioinformatics methodologies, resulting in the construction of the lncRNA-miRNA-mRNA network. Among these, lncRNA (MIR17HG) showed a high correlation with PANoptosis. Bibliometric methods were employed to collect literature data on PANoptosis, and its components were inferred. PCR and Western Blotting experiments confirmed that lncRNA MIR17HG is related to PANoptosis in osteoblasts during inflammation. CONCLUSIONS: Our data suggest that TNF-α-induced inhibition of osteogenic differentiation and PANoptosis in MC3T3-E1 osteoblasts is associated with MIR17HG. These findings highlight the critical role of MIR17HG in the interplay between inflammation, PANoptosis, and osteogenic differentiation, suggesting potential therapeutic targets for conditions involving impaired bone formation and inflammatory responses.


Subject(s)
Cell Differentiation , Gene Regulatory Networks , Osteogenesis , RNA, Competitive Endogenous , RNA, Long Noncoding , Tumor Necrosis Factor-alpha , Animals , Humans , Mice , Apoptosis/genetics , Cell Differentiation/genetics , Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Inflammation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/genetics , RNA, Competitive Endogenous/genetics , RNA, Competitive Endogenous/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125811

ABSTRACT

Advanced glycated end products (AGEs) are cytotoxic compounds that are mainly increased in diabetes mellitus (DM), kidney failure, inflammation, and in response to the ingestion of AGE-rich diets. AGEs can also impair glycemic homeostasis by decreasing the expression of the Slc2a4 (solute carrier family 2 member 4) gene and its GLUT4 (solute carrier family 2, facilitated glucose transporter member 4) protein in muscle. However, the mechanisms underlying AGE's effect on adipocytes have not been demonstrated yet. This study investigated the effects of AGEs upon Slc2a4/GLUT4 expression in 3T3-L1 adipocytes, as well as the potential role of NFKB (nuclear factor NF-kappa-B) activity in the effects observed. Adipocytes were cultured in the presence of control albumin (CA) or advanced glycated albumin (GA) at concentrations of 0.4, 3.6, and 5.4 mg/mL for 24 h or 72 h. Slc2a4, Rela, and Nfkb1mRNAs were measured by RT-qPCR, GLUT4, IKKA/B, and p50/p65 NFKB subunits using Western blotting, and p50/p65 binding into the Slc2a4 promoter was analyzed by chromatin immunoprecipitation (ChIP) assay. GA at 0.4 mg/mL increased Slc2a4/GLUT4 expression after 24 h and 72 h (from 50% to 100%), but at 5.4 mg/mL, Slc2a4/GLUT4 expression decreased at 72 h (by 50%). Rela and Nfkb1 expression increased after 24 h at all concentrations, but this effect was not observed at 72 h. Furthermore, 5.4 mg/mL of GA increased the p50/p65 nuclear content and binding into Slc2a4 at 72 h. In summary, this study reveals AGE-induced and NFKB-mediated repression of Slc2a4/GLUT4 expression. This can compromise the adipocyte glucose utilization, contributing not only to the worsening of glycemic control in DM subjects but also the impairment of glycemic homeostasis in non-DM subjects under the high intake of AGE-rich foods.


Subject(s)
3T3-L1 Cells , Adipocytes , Glucose Transporter Type 4 , Glycation End Products, Advanced , Transcription Factor RelA , Animals , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 4/genetics , Mice , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/pharmacology , Adipocytes/metabolism , Adipocytes/drug effects , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , NF-kappa B/metabolism , Gene Expression Regulation/drug effects , Promoter Regions, Genetic , NF-kappa B p50 Subunit/metabolism , NF-kappa B p50 Subunit/genetics
10.
Cells ; 13(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39120336

ABSTRACT

Platelet-rich fibrin (PRF) is prepared by spontaneous coagulation of fractionated blood. When squeezed between two plates, PRF is separated into solid PRF membranes and a liquid exudate, the PRF serum. The question arises regarding how much the overall activity remains in the PRF membranes and what is discarded into the PRF serum. To this end, we have exposed gingival fibroblasts to lysates prepared from PRF membranes and PRF serum, followed by bulk RNA sequencing. A total of 268 up- and 136 down-regulated genes in gingival fibroblasts exposed to PRF membrane lysates were significantly regulated under the premise of a minimum log2 with 2.5-fold change and a minus log10 significance level of two, respectively. PRF serum only caused 62 up- and 32 down-regulated genes under these conditions. Among the 46 commonly up-regulated genes were CXCL1, CXCL5, CXCL6, CXCL8, IL33, IL6, and PTGS2/COX2, stanniocalcin-1-all linked to an inflammatory response. PRF membrane lysates further increased chemokines CCL2, CCL7, CXCL2, CXCL3, and IL1R1, IL1RL1, and IL1RN, as well as the paracrine factors IL11, LIF, IGF1, BMP2, BMP6, FGF2, and CCN2/CTGF, and all hyaluronan synthases. On the other hand, PRF serum increased DKK1. The genes commonly down-regulated by PRF membrane lysates and PRF serum included interferon-induced protein with tetratricopeptide repeats (IFIT1, IFIT2, IFIT3) and odd-skipped-related transcription factors (OSR1 and OSR2), as well as FGF18 and GDF15, respectively. Taken together, PRF membrane lysates, compared to PRF serum, cause a more complex response in gingival fibroblasts, but each increased chemokine expression in gingival fibroblasts.


Subject(s)
Fibroblasts , Gingiva , Platelet-Rich Fibrin , Humans , Fibroblasts/metabolism , Fibroblasts/drug effects , Gingiva/cytology , Gingiva/metabolism , Platelet-Rich Fibrin/metabolism , Gene Expression Regulation/drug effects
11.
Nutrients ; 16(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125278

ABSTRACT

(1) Background: We examined the effect of the acute administration of olive oil (EVOO), linseed oil (GLO), soybean oil (SO), and palm oil (PO) on gastric motility and appetite in rats. (2) Methods: We assessed food intake, gastric retention (GR), and gene expression in all groups. (3) Results: Both EVOO and GLO were found to enhance the rate of stomach retention, leading to a decrease in hunger. On the other hand, the reduction in food intake caused by SO was accompanied by delayed effects on stomach retention. PO caused an alteration in the mRNA expression of NPY, POMC, and CART. Although PO increased stomach retention after 180 min, it did not affect food intake. It was subsequently verified that the absence of an autonomic reaction did not nullify the influence of EVOO in reducing food consumption. Moreover, in the absence of parasympathetic responses, animals that received PO exhibited a significant decrease in food consumption, probably mediated by lower NPY expression. (4) Conclusions: This study discovered that different oils induce various effects on parameters related to food consumption. Specifically, EVOO reduces food consumption primarily through its impact on the gastrointestinal tract, making it a recommended adjunct for weight loss. Conversely, the intake of PO limits food consumption in the absence of an autonomic reaction, but it is not advised due to its contribution to the development of cardiometabolic disorders.


Subject(s)
Appetite Regulation , Hypothalamus , Neuropeptide Y , Olive Oil , Palm Oil , Soybean Oil , Vagus Nerve , Animals , Vagus Nerve/drug effects , Vagus Nerve/physiology , Hypothalamus/metabolism , Hypothalamus/drug effects , Male , Olive Oil/pharmacology , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Palm Oil/pharmacology , Appetite Regulation/drug effects , Soybean Oil/administration & dosage , Soybean Oil/pharmacology , Rats, Wistar , Linseed Oil/pharmacology , Rats , Eating/drug effects , Plant Oils/pharmacology , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Gastrointestinal Motility/drug effects , Gene Expression Regulation/drug effects , RNA, Messenger/metabolism , RNA, Messenger/genetics
12.
Nutrients ; 16(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39125380

ABSTRACT

BACKGROUND: Excessive fluoride exposure induces skeletal fluorosis, but the specific mechanism responsible is still unclear. Therefore, this study aimed to identify the pathogenesis of fluoride-induced bone injuries. METHODS: We systematically searched fluoride-induced bone injury-related genes from five databases. Then, these genes were subjected to enrichment analyses. A TF (transcription factor)-mRNA-miRNA network and protein-protein interaction (PPI) network were constructed using Cytoscape, and the Human Protein Atlas (HPA) database was used to screen the expression of key proteins. The candidate pharmacological targets were predicted using the Drug Signature Database. RESULTS: A total of 85 studies were included in this study, and 112 osteoblast-, 35 osteoclast-, and 41 chondrocyte-related differential expression genes (DEGs) were identified. Functional enrichment analyses showed that the Atf4, Bcl2, Col1a1, Fgf21, Fgfr1 and Il6 genes were significantly enriched in the PI3K-Akt signaling pathway of osteoblasts, Mmp9 and Mmp13 genes were enriched in the IL-17 signaling pathway of osteoclasts, and Bmp2 and Bmp7 genes were enriched in the TGF-beta signaling pathway of chondrocytes. With the use of the TF-mRNA-miRNA network, the Col1a1, Bcl2, Fgfr1, Mmp9, Mmp13, Bmp2, and Bmp7 genes were identified as the key regulatory factors. Selenium methyl cysteine, CGS-27023A, and calcium phosphate were predicted to be the potential drugs for skeletal fluorosis. CONCLUSIONS: These results suggested that the PI3K-Akt signaling pathway being involved in the apoptosis of osteoblasts, with the IL-17 and the TGF-beta signaling pathways being involved in the inflammation of osteoclasts and chondrocytes in fluoride-induced bone injuries.


Subject(s)
Apoptosis , Fluorides , Inflammation , Osteoblasts , Signal Transduction , Humans , Fluorides/adverse effects , Apoptosis/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Inflammation/chemically induced , Signal Transduction/drug effects , MicroRNAs/metabolism , MicroRNAs/genetics , Osteoclasts/drug effects , Osteoclasts/metabolism , Chondrocytes/drug effects , Chondrocytes/metabolism , Protein Interaction Maps , RNA, Messenger/metabolism , RNA, Messenger/genetics , Gene Regulatory Networks , Gene Expression Regulation/drug effects , Bone Diseases/chemically induced , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125611

ABSTRACT

Sexual dimorphism among mammals includes variations in the pain threshold. These differences are influenced by hormonal fluctuations in females during the estrous and menstrual cycles of rodents and humans, respectively. These physiological conditions display various phases, including proestrus and diestrus in rodents and follicular and luteal phases in humans, distinctly characterized by varying estrogen levels. In this study, we evaluated the capsaicin responses in male and female mice at different estrous cycle phases, using two murine acute pain models. Our findings indicate that the capsaicin-induced pain threshold was lower in the proestrus phase than in the other three phases in both pain assays. We also found that male mice exhibited a higher pain threshold than females in the proestrus phase, although it was similar to females in the other cycle phases. We also assessed the mRNA and protein levels of TRPV1 in the dorsal root and trigeminal ganglia of mice. Our results showed higher TRPV1 protein levels during proestrus compared to diestrus and male mice. Unexpectedly, we observed that the diestrus phase was associated with higher TRPV1 mRNA levels than those in both proestrus and male mice. These results underscore the hormonal influence on TRPV1 expression regulation and highlight the role of sex steroids in capsaicin-induced pain.


Subject(s)
Capsaicin , Pain , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Capsaicin/pharmacology , Male , Female , Mice , Pain/metabolism , Pain/genetics , Gonadal Steroid Hormones/metabolism , Estrous Cycle/drug effects , Pain Threshold/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/drug effects , Gene Expression Regulation/drug effects , Sex Characteristics , RNA, Messenger/metabolism , RNA, Messenger/genetics
14.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125657

ABSTRACT

Well-controlled type 1 diabetes (T1DM) is characterized by inflammation and endothelial dysfunction, thus constituting a suitable model of subclinical cardiovascular disease (CVD). miR-199b-5p overexpression in murine CVD has shown proatherosclerotic effects. We hypothesized that miR-199b-5p would be overexpressed in subclinical CVD yet downregulated following metformin therapy. Inflammatory and vascular markers were measured in 29 individuals with T1DM and 20 matched healthy controls (HCs). miR-199b-5p expression in CFU-Hill's colonies was analyzed from each study group, and correlations with inflammatory/vascular health indices were evaluated. Significant upregulation of miR-199b-5p was observed in T1DM, which was significantly downregulated by metformin. miR-199b-5p correlated positively with vascular endothelial growth factor-D and c-reactive protein (CRP: nonsignificant). ROC analysis determined miR-199b-5p to define subclinical CVD by discriminating between HCs and T1DM individuals. ROC analyses of HbA1c and CRP showed that the upregulation of miR-199b-5p in T1DM individuals defined subclinical CVD at HbA1c > 44.25 mmol and CRP > 4.35 × 106 pg/mL. Ingenuity pathway analysis predicted miR-199b-5p to inhibit the target genes SIRT1, ETS1, and JAG1. Metformin was predicted to downregulate miR-199b-5p via NFATC2 and STAT3 and reverse its downstream effects. This study validated the antiangiogenic properties of miR-199b-5p and substantiated miR-199b-5p overexpression as a biomarker of subclinical CVD. The downregulation of miR-199b-5p by metformin confirmed its cardio-protective effect.


Subject(s)
Cardiovascular Diseases , Metformin , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Humans , Male , Female , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Adult , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Inflammation/genetics , Inflammation/drug therapy , Inflammation/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics , C-Reactive Protein/metabolism , C-Reactive Protein/genetics , Middle Aged , Gene Expression Regulation/drug effects , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Biomarkers , Case-Control Studies
15.
PLoS One ; 19(8): e0304183, 2024.
Article in English | MEDLINE | ID: mdl-39088439

ABSTRACT

Expression of the androgen receptor is key to the response of cells and tissues to androgenic steroids, such as testosterone or dihydrotestosterone, as well as impacting the benefit of hormone-dependent therapies for endocrine diseases and hormone-dependent cancers. However, the mechanisms controlling androgen receptor expression are not fully understood, limiting our ability to effectively promote or inhibit androgenic signalling therapeutically. An autoregulatory loop has been described in which androgen receptor may repress its own expression in the presence of hormone, although the molecular mechanisms are not fully understood. In this work, we elucidate the mechanisms of autoregulation and demonstrate, for the first time, that a similar repression of the AR gene is facilitated by the progesterone receptor. We show that the progesterone receptor, like the androgen receptor binds to response elements within the AR gene to effect transcriptional repression in response to hormone treatment. Mechanistically, this repression involves hormone-dependent histone deacetylation within the AR 5'UTR region and looping between sequences in intron 2 and the transcription start site (TSS). This novel pathway controlling AR expression in response to hormone stimulation may have important implications for understanding cell or tissue selective receptor signalling.


Subject(s)
Gene Expression Regulation , Receptors, Androgen , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Humans , Gene Expression Regulation/drug effects , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , 5' Untranslated Regions , Response Elements , Cell Line, Tumor , Acetylation , Transcription, Genetic/drug effects
16.
Vet Q ; 44(1): 1-11, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39086189

ABSTRACT

Mare endometrosis is a major reproductive problem associated with low fertility and is characterized by persistent inflammation, TGFß-1 signaling, and consequently, extracellular matrix deposition, which compromises endometrial glands. Mesenchymal stem cell-based products (MSCs), such as extracellular vesicles (EVs), have gained attention due to the regulatory effects exerted by their miRNA cargo. Here, we evaluated the impact of preconditioning equine adipose mesenchymal stem cells with TGFß-1 for short or long periods on the anti-fibrotic properties of secreted extracellular vesicles. MSCs were isolated from six healthy horses and exposed to TGFß-1 for 4, 24, and 0 h. The expression of anti-fibrotic and pro-fibrotic miRNAs and mRNAs in treated cells and miRNAs in the cargo of secreted extracellular vesicles was measured. The resulting EVs were added for 48 h to endometrial stromal cells previously induced to a fibrotic status. The expression of anti-fibrotic and pro-fibrotic genes and miRNAs was evaluated in said cells using qPCR and next-generation sequencing. Preconditioning MSCs with TGFß-1 for 4 h enriched the anti-fibrotic miRNAs (mir29c, mir145, and mir200) in cells and EVs. Conversely, preconditioning the cells for 24 h leads to a pro-fibrotic phenotype overexpressing mir192 and mir433. This finding might have implications for developing an EV-based protocol to treat endometrial fibrosis in mares.


Subject(s)
Endometrium , Extracellular Vesicles , Fibrosis , Mesenchymal Stem Cells , MicroRNAs , Transforming Growth Factor beta1 , Animals , Horses , Female , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Extracellular Vesicles/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Endometrium/metabolism , Endometrium/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Stromal Cells/metabolism , Stromal Cells/drug effects , Horse Diseases , Gene Expression Regulation/drug effects , Endometriosis/veterinary , Endometriosis/metabolism , Endometriosis/genetics
17.
Front Immunol ; 15: 1410150, 2024.
Article in English | MEDLINE | ID: mdl-38947331

ABSTRACT

The recent trend of global warming poses a significant threat to ecosystems worldwide. This global climate change has also impacted the pollution levels in aquatic ecosystems, subsequently affecting human health. To address these issues, an experiment was conducted to investigate the mitigating effects of iron nanoparticles (Fe-NPs) on arsenic and ammonia toxicity as well as high temperature stress (As+NH3+T). Fe-NPs were biologically synthesized using fish waste and incorporated into feed formulations at 10, 15, and 20 mg kg-1 diet. A total of 12 treatments were designed in triplicate following a completely randomized design involving 540 fish. Fe-NPs at 15 mg kg-1 diet notably reduced the cortisol levels in fish exposed to multiple stressors. The gene expressions of HSP 70, DNA damage-inducible protein (DDIP), and DNA damage were upregulated by stressors (As+NH3+T) and downregulated by Fe-NPs. Apoptotic genes (Cas 3a and 3b) and detoxifying genes (CYP 450), metallothionein (MT), and inducible nitric oxide synthase (iNOS) were downregulated by Fe-NPs at 15 mg kg-1 diet in fish subjected to As+NH3+T stress. Immune-related genes such as tumor necrosis factor (TNFα), immunoglobulin (Ig), and interleukin (IL) were upregulated by Fe-NPs, indicating enhanced immunity in fish under As+NH3+T stress. Conversely, Toll-like receptor (TLR) expression was notably downregulated by Fe-NPs at 15 mg kg-1 diet in fish under As+NH3+T stress. Immunological attributes such as nitro blue tetrazolium chloride, total protein, albumin, globulin, A:G ratio, and myeloperoxidase (MPO) were improved by dietary Fe-NPs at 15 mg kg-1 diet in fish, regardless of stressors. The antioxidant genes (CAT, SOD, and GPx) were also strengthened by Fe-NPs in fish. Genes associated with growth performance, such as growth hormone regulator (GHR1 and GHRß), growth hormone (GH), and insulin-like growth factor (IGF 1X and IGF 2X), were upregulated, enhancing fish growth under stress, while SMT and MYST were downregulated by Fe-NPs in the diet. Various growth performance indicators were improved by dietary Fe-NPs at 15 mg kg-1 diet. Notably, Fe-NPs also enhanced arsenic detoxification and reduced the cumulative mortality after a bacterial infection. In conclusion, this study highlights that dietary Fe-NPs can effectively mitigate arsenic and ammonia toxicity as well as high temperature stress by modulating gene expression in fish.


Subject(s)
Fishes , Gene Expression Regulation , Iron , Stress, Physiological , Animals , Gene Expression Regulation/drug effects , Iron/metabolism , Fishes/immunology , Stress, Physiological/immunology , Stress, Physiological/drug effects , Metal Nanoparticles , Arsenic/toxicity
18.
J Biochem Mol Toxicol ; 38(7): e23764, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38963172

ABSTRACT

Obesity is an established risk factor for numerous malignancies, although it remains uncertain whether the disease itself or weight-loss drugs are responsible for a greater predisposition to cancer. The objective of the current study was to determine the impact of dulaglutide on genetic and epigenetic DNA damage caused by obesity, which is a crucial factor in the development of cancer. Mice were administered a low-fat or high-fat diet for 12 weeks, followed by a 5-week treatment with dulaglutide. Following that, modifications of the DNA bases were examined using the comet assay. To clarify the underlying molecular mechanisms, oxidized and methylated DNA bases, changes in the redox status, levels of inflammatory cytokines, and the expression levels of some DNA repair genes were evaluated. Animals fed a high-fat diet exhibited increased body weights, elevated DNA damage, oxidation of DNA bases, and DNA hypermethylation. In addition, obese mice showed altered inflammatory responses, redox imbalances, and repair gene expressions. The findings demonstrated that dulaglutide does not exhibit genotoxicity in the investigated conditions. Following dulaglutide administration, animals fed a high-fat diet demonstrated low DNA damage, less oxidation and methylation of DNA bases, restored redox balance, and improved inflammatory responses. In addition, dulaglutide treatment restored the upregulated DNMT1, Ogg1, and p53 gene expression. Overall, dulaglutide effectively maintains DNA integrity in obese animals. It reduces oxidative DNA damage and hypermethylation by restoring redox balance, modulating inflammatory responses, and recovering altered gene expressions. These findings demonstrate dulaglutide's expediency in treating obesity and its associated complications.


Subject(s)
DNA Damage , DNA Methylation , DNA Repair , Diet, High-Fat , Glucagon-Like Peptides , Immunoglobulin Fc Fragments , Oxidation-Reduction , Recombinant Fusion Proteins , Animals , Glucagon-Like Peptides/analogs & derivatives , Glucagon-Like Peptides/pharmacology , DNA Methylation/drug effects , Immunoglobulin Fc Fragments/pharmacology , DNA Damage/drug effects , Mice , DNA Repair/drug effects , Diet, High-Fat/adverse effects , Recombinant Fusion Proteins/pharmacology , Male , Oxidation-Reduction/drug effects , Inflammation/metabolism , Inflammation/genetics , Oxidative Stress/drug effects , Obesity/metabolism , Obesity/drug therapy , Obesity/genetics , Gene Expression Regulation/drug effects , Mice, Inbred C57BL
19.
J Diabetes Res ; 2024: 8555211, 2024.
Article in English | MEDLINE | ID: mdl-39022651

ABSTRACT

We have previously identified a parasite-derived peptide, FhHDM-1, that prevented the progression of diabetes in nonobese diabetic (NOD) mice. Disease prevention was mediated by the activation of the PI3K/Akt pathway to promote ß-cell survival and metabolism without inducing proliferation. To determine the molecular mechanisms driving the antidiabetogenic effects of FhHDM-1, miRNA:mRNA interactions and in silico predictions of the gene networks were characterised in ß-cells, which were exposed to the proinflammatory cytokines that mediate ß-cell destruction in Type 1 diabetes (T1D), in the presence and absence of FhHDM-1. The predicted gene targets of miRNAs differentially regulated by FhHDM-1 mapped to the biological pathways that regulate ß-cell biology. Six miRNAs were identified as important nodes in the regulation of PI3K/Akt signaling. Additionally, IGF-2 was identified as a miRNA gene target that mediated the beneficial effects of FhHDM-1 on ß-cells. The findings provide a putative mechanism by which FhHDM-1 positively impacts ß-cells to permanently prevent diabetes. As ß-cell death/dysfunction underlies diabetes development, FhHDM-1 opens new therapeutic avenues.


Subject(s)
Apoptosis , Cytokines , Insulin-Secreting Cells , MicroRNAs , Signal Transduction , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Apoptosis/drug effects , Signal Transduction/drug effects , Cytokines/metabolism , Mice , Mice, Inbred NOD , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Diabetes Mellitus, Type 1/metabolism , Gene Expression Regulation/drug effects
20.
BMC Vet Res ; 20(1): 321, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026262

ABSTRACT

BACKGROUND: Identifying alternative sustainable feed sources with high nutritional values is crucial for the future of environmentally and socially responsible aquaculture. In this regard, microalgae have been proven to have positive effects on fish health, which overwhelmed our interest in this study. METHODS: Pediastrum boryanum (P. boryanum) was incorporated into Nile tilapia feed at concentrations of 0, 0.75, and 1.5 mg/kg, as control, PbExt0.75, and PbExt1.5 groups to assess its effects on growth and biochemical indices, oxidant/antioxidant activities, immune and stress-related gene expression, and intestinal morphology. RESULTS: After 8 weeks, fish fed P. boryanum supplemented feed exhibited significant increases in final weight, length, condition factor, body weight gain, and specific growth rate, while the spleen-somatic index (SSI) and hepatosomatic index (HSI) showed no significant differences compared to the control group. Dietary P. boryanum supplementation also enhanced IgM levels and lysozyme activity, along with no marked effect on markers of liver function enzymes (alanine aminotransferase/ALT and aspartate aminotransferase/AST) or protein status (total protein and albumin). Furthermore, P. boryanum addition increased the activity of superoxide dismutase (SOD), catalase (CAT), and reduced glutathione (GSH) enzymes, highlighting its antioxidant potential, whereas malondialdehyde (MDA) concentrations showed no significant differences among the groups. Gene expression analysis revealed that tumor necrosis factor-α (TNF-α), interleukin-10 (IL-10), and transforming growth factor-ß1 (TGF-ß1) expression notably increased in groups fed P. boryanum containing feed, while no significant difference was observed in hepatic Heat Shock Protein 70 (HSP70) mRNA expression. Histopathological examination revealed no adverse effects of P. boryanum supplementation on the liver, spleen, or intestinal tissues. Villous height and villous surface area were notably increased in the high P. boryanum supplementation group, suggesting improved intestinal integrity and nutrient absorption. CONCLUSION: Dietary P. boryanum supplementation can potentially improve growth performance, immune response, antioxidant status, and intestinal health of Nile tilapia, making it a promising candidate for sustainable aquaculture.


Subject(s)
Animal Feed , Cichlids , Diet , Dietary Supplements , Microalgae , Animals , Cichlids/immunology , Cichlids/growth & development , Animal Feed/analysis , Diet/veterinary , Aquaculture , Antioxidants/metabolism , Gene Expression Regulation/drug effects , Plant Extracts/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL