Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273.283
Filter
1.
Iran J Allergy Asthma Immunol ; 23(2): 182-196, 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38822513

ABSTRACT

Multiple sclerosis (MS) is an autoimmune neurodegenerative disease and has adverse implications. The exact mechanism of its pathogenesis is not fully understood and remains to be elucidated. In the current study we aimed to identify key genes that can serve as potential biomarkers and therapeutic targets for MS and shed light on pathogenesis mechanisms involved in MS. We analyzed a gene expression dataset (GES21942) and found 266 differentially expressed genes (DEGs) including 183 upregulated and 83 downregulated genes in MS patients compared to controls. Then we conducted pathway enrichment on DEGs and selected the top enriched pathway i.e., B cell receptor signaling pathway, and 5 genes of this pathway (CR2, BLK, BLNK, RASGRP3, and KRAS) for further investigation in our clinical samples. We recruited 50 MS patients and 50 controls and assessed the expression of selected genes in the circulation of patients versus controls. Expression of CR2, BLK, BLNK, and RASGRP3 were significantly higher in MS cases compared with controls. There was no significant difference in expression of KRAS between patients and controls. All of the selected genes with differential expression had noticeable diagnostic power and CR2 was the most robust gene in differentiating MS cases from controls. Additionally, a combination of genes resulted in enhanced diagnostic power. Collectively our results suggest that the B cell receptor signaling pathway and the selected genes from this pathway may be implicated in the pathogenesis of MS and each of these genes can be considered as potential diagnostic biomarkers and therapeutic targets.


Subject(s)
Multiple Sclerosis , Receptors, Antigen, B-Cell , Signal Transduction , Humans , Signal Transduction/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/blood , Female , Male , Adult , Receptors, Antigen, B-Cell/genetics , Gene Expression Profiling , Case-Control Studies , Biomarkers , Middle Aged , Gene Expression Regulation
2.
Mol Biol Rep ; 51(1): 710, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824241

ABSTRACT

BACKGROUND: Circular RNA (circRNA) is a key player in regulating the multidirectional differentiation of stem cells. Previous research by our group found that the blue light-emitting diode (LED) had a promoting effect on the osteogenic/odontogenic differentiation of human stem cells from apical papilla (SCAPs). This research aimed to investigate the differential expression of circRNAs during the osteogenic/odontogenic differentiation of SCAPs regulated by blue LED. MATERIALS AND METHODS: SCAPs were divided into the irradiation group (4 J/cm2) and the control group (0 J/cm2), and cultivated in an osteogenic/odontogenic environment. The differentially expressed circRNAs during osteogenic/odontogenic differentiation of SCAPs promoted by blue LED were detected by high-throughput sequencing, and preliminarily verified by qRT-PCR. Functional prediction of these circRNAs was performed using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) and the circRNA-miRNA-mRNA networks were also constructed. RESULTS: It showed 301 circRNAs were differentially expressed. GO and KEGG analyses suggested that these circRNAs were associated with some signaling pathways related to osteogenic/odontogenic differentiation. And the circRNA-miRNA-mRNA networks were also successfully constructed. CONCLUSION: CircRNAs were involved in the osteogenic/odontogenic differentiation of SCAPs promoted by blue LED. In this biological process, circRNA-miRNA-mRNA networks served an important purpose, and circRNAs regulated this process through certain signaling pathways.


Subject(s)
Cell Differentiation , Dental Papilla , Light , Odontogenesis , Osteogenesis , RNA, Circular , Stem Cells , RNA, Circular/genetics , RNA, Circular/metabolism , Humans , Osteogenesis/genetics , Cell Differentiation/genetics , Stem Cells/metabolism , Stem Cells/cytology , Odontogenesis/genetics , Dental Papilla/cytology , Dental Papilla/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Ontology , Cells, Cultured , Gene Expression Profiling/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing/methods , Gene Expression Regulation/radiation effects , Blue Light
3.
Mol Biol Rep ; 51(1): 712, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824221

ABSTRACT

INTRODUCTION: Coronary artery disease (CAD) in young adults can have devastating consequences. The cardiac developmental gene MEIS1 plays important roles in vascular networks and heart development. This gene effects on the regeneration capacity of the heart. Considering role of MEIS1 in cardiac tissue development and the progression of myocardial infarction this study investigated the expression levels of the MEIS1, HIRA, and Myocardin genes in premature CAD patients compared to healthy subjects and evaluated the relationships between these genes and possible inflammatory factors. METHODS AND RESULTS: The study conducted a case-control design involving 35 CAD patients and 35 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were collected, and gene expression analysis was performed using real-time PCR. Compared with control group, the number of PBMCs in the CAD group exhibited greater MEIS1 and HIRA gene expression, with fold changes of 2.45 and 3.6. The expression of MEIS1 exhibited a negative correlation with IL-10 (r= -0.312) expression and positive correlation with Interleukin (IL)-6 (r = 0.415) and tumor necrosis factor (TNF)-α (r = 0.534) gene expression. Moreover, there was an inverse correlation between the gene expression of HIRA and that of IL-10 (r= -0.326), and a positive correlation was revealed between the expression of this gene and that of the IL-6 (r = 0.453) and TNF-α (r = 0.572) genes. CONCLUSION: This research demonstrated a disparity in expression levels of MEIS1, HIRA, and Myocardin, between CAD and healthy subjects. The results showed that, MEIS1 and HIRA play significant roles in regulating the synthesis of proinflammatory cytokines, namely, TNF-α and IL-6.


Subject(s)
Coronary Artery Disease , Myeloid Ecotropic Viral Integration Site 1 Protein , Nuclear Proteins , Trans-Activators , Adult , Female , Humans , Male , Middle Aged , Case-Control Studies , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Coronary Artery Disease/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , Interleukin-10/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Leukocytes, Mononuclear/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
Mol Biol Rep ; 51(1): 709, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824265

ABSTRACT

BACKGROUND: Cystatin is a protease inhibitor that also regulates genes expression linked to inflammation and plays a role in defense and regulation. METHODS AND RESULTS: Cystatin 10 (Smcys10) was cloned from Scophthalmus maximus and encodes a 145 amino acid polypeptide. The results of qRT-PCR showed that Smcys10 exhibited tissue-specific expression patterns, and its expression was significantly higher in the skin than in other tissues. The expression level of Smcys10 was significantly different in the skin, gill, head kidney, spleen and macrophages after Vibrio anguillarum infection, indicating that Smcys10 may play an important role in resistance to V. anguillarum infection. The recombinant Smcys10 protein showed binding and agglutinating activity in a Ca2+-dependent manner against bacteria. rSmcys10 treatment upregulated the expression of IL-10, TNF-α and TGF-ß in macrophages of turbot and hindered the release of lactate dehydrogenase (LDH) from macrophages after V. anguillarum infection, which confirmed that rSmcys10 reduced the damage to macrophages by V. anguillarum. The NF-κB pathway was suppressed by Smcys10, as demonstrated by dual-luciferase analysis. CONCLUSIONS: These results indicated that Smcys10 is involved in the host antibacterial immune response.


Subject(s)
Cystatins , Fish Diseases , Fish Proteins , Flatfishes , Macrophages , Vibrio , Animals , Flatfishes/immunology , Flatfishes/genetics , Flatfishes/metabolism , Vibrio/pathogenicity , Cystatins/genetics , Cystatins/metabolism , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/immunology , Macrophages/metabolism , Macrophages/immunology , Fish Diseases/immunology , Fish Diseases/genetics , Fish Diseases/microbiology , Vibrio Infections/immunology , Vibrio Infections/veterinary , Vibrio Infections/genetics , NF-kappa B/metabolism , Cloning, Molecular/methods , Gene Expression Regulation
5.
Nat Commun ; 15(1): 4839, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844462

ABSTRACT

Comparative genomics has revealed the rapid expansion of multiple gene families involved in immunity. Members within each gene family often evolved distinct roles in immunity. However, less is known about the evolution of their epigenome and cis-regulation. Here we systematically profile the epigenome of the recently expanded murine Ly49 gene family that mainly encode either inhibitory or activating surface receptors on natural killer cells. We identify a set of cis-regulatory elements (CREs) for activating Ly49 genes. In addition, we show that in mice, inhibitory and activating Ly49 genes are regulated by two separate sets of proximal CREs, likely resulting from lineage-specific losses of CRE activity. Furthermore, we find that some Ly49 genes are cross-regulated by the CREs of other Ly49 genes, suggesting that the Ly49 family has begun to evolve a concerted cis-regulatory mechanism. Collectively, we demonstrate the different modes of cis-regulatory evolution for a rapidly expanding gene family.


Subject(s)
Evolution, Molecular , Multigene Family , NK Cell Lectin-Like Receptor Subfamily A , Animals , Mice , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Gene Expression Regulation , Killer Cells, Natural/immunology , Mice, Inbred C57BL
6.
Nat Commun ; 15(1): 4894, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38849338

ABSTRACT

Synthetic biology applications require finely tuned gene expression, often mediated by synthetic transcription factors (sTFs) compatible with the human genome and transcriptional regulation mechanisms. While various DNA-binding and activation domains have been developed for different applications, advanced artificially controllable sTFs with improved regulatory capabilities are required for increasingly sophisticated applications. Here, in mammalian cells and mice, we validate the transactivator function and homo-/heterodimerization activity of the plant-derived phytochrome chaperone proteins, FHY1 and FHL. Our results demonstrate that FHY1/FHL form a photosensing transcriptional regulation complex (PTRC) through interaction with the phytochrome, ΔPhyA, that can toggle between active and inactive states through exposure to red or far-red light, respectively. Exploiting this capability, we develop a light-switchable platform that allows for orthogonal, modular, and tunable control of gene transcription, and incorporate it into a PTRC-controlled CRISPRa system (PTRCdcas) to modulate endogenous gene expression. We then integrate the PTRC with small molecule- or blue light-inducible regulatory modules to construct a variety of highly tunable systems that allow rapid and reversible control of transcriptional regulation in vitro and in vivo. Validation and deployment of these plant-derived phytochrome chaperone proteins in a PTRC platform have produced a versatile, powerful tool for advanced research and biomedical engineering applications.


Subject(s)
Light , Molecular Chaperones , Phytochrome , Animals , Humans , Mice , Phytochrome/metabolism , Phytochrome/genetics , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Gene Expression Regulation/radiation effects , Transcription, Genetic/radiation effects , HEK293 Cells , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Arabidopsis/genetics , Arabidopsis/metabolism , Plant Proteins/metabolism , Plant Proteins/genetics
7.
Nat Commun ; 15(1): 4890, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849352

ABSTRACT

The human brain has been implicated in the pathogenesis of several complex diseases. Taking advantage of single-cell techniques, genome-wide association studies (GWAS) have taken it a step further and revealed brain cell-type-specific functions for disease loci. However, genetic causal associations inferred by Mendelian randomization (MR) studies usually include all instrumental variables from GWAS, which hampers the understanding of cell-specific causality. Here, we developed an analytical framework, Cell-Stratified MR (csMR), to investigate cell-stratified causality through colocalizing GWAS signals with single-cell eQTL from different brain cells. By applying to obesity-related traits, our results demonstrate the cell-type-specific effects of GWAS variants on gene expression, and indicate the benefits of csMR to identify cell-type-specific causal effect that is often hidden from bulk analyses. We also found csMR valuable to reveal distinct causal pathways between different obesity indicators. These findings suggest the value of our approach to prioritize target cells for extending genetic causation studies.


Subject(s)
Brain , Genome-Wide Association Study , Mendelian Randomization Analysis , Obesity , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Humans , Obesity/genetics , Obesity/metabolism , Brain/metabolism , Single-Cell Analysis/methods , Genetic Predisposition to Disease/genetics , Causality , Gene Expression Regulation , Gene Expression/genetics
8.
Genome Biol ; 25(1): 144, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822397

ABSTRACT

BACKGROUND: Variation in X chromosome inactivation (XCI) in human-induced pluripotent stem cells (hiPSCs) can impact their ability to model biological sex biases. The gene-wise landscape of X chromosome gene dosage remains unresolved in female hiPSCs. To characterize patterns of de-repression and escape from inactivation, we performed a systematic survey of allele specific expression in 165 female hiPSC lines. RESULTS: XCI erosion is non-random and primarily affects genes that escape XCI in human tissues. Individual genes and cell lines vary in the frequency and degree of de-repression. Bi-allelic expression increases gradually after modest decrease of XIST in cultures, whose loss is commonly used to mark lines with eroded XCI. We identify three clusters of female lines at different stages of XCI. Increased XCI erosion amplifies female-biased expression at hypomethylated sites and regions normally occupied by repressive histone marks, lowering male-biased differences in the X chromosome. In autosomes, erosion modifies sex differences in a dose-dependent way. Male-biased genes are enriched for hypermethylated regions, and de-repression of XIST-bound autosomal genes in female lines attenuates normal male-biased gene expression in eroded lines. XCI erosion can compensate for a dominant loss of function effect in several disease genes. CONCLUSIONS: We present a comprehensive view of X chromosome gene dosage in hiPSCs and implicate a direct mechanism for XCI erosion in regulating autosomal gene expression in trans. The uncommon and variable reactivation of X chromosome genes in female hiPSCs can provide insight into X chromosome's role in regulating gene expression and sex differences in humans.


Subject(s)
Chromosomes, Human, X , Induced Pluripotent Stem Cells , RNA, Long Noncoding , X Chromosome Inactivation , Humans , Induced Pluripotent Stem Cells/metabolism , Female , Chromosomes, Human, X/genetics , Male , RNA, Long Noncoding/genetics , Alleles , Gene Expression Regulation , DNA Methylation
9.
Proc Natl Acad Sci U S A ; 121(24): e2321267121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838014

ABSTRACT

Mitochondria perform an array of functions, many of which involve interactions with gene products encoded by the nucleus. These mitochondrial functions, particularly those involving energy production, can be expected to differ between sexes and across ages. Here, we measured mitochondrial effects on sex- and age-specific gene expression in parental and reciprocal F1 hybrids between allopatric populations of Tigriopus californicus with over 20% mitochondrial DNA divergence. Because the species lacks sex chromosomes, sex-biased mitochondrial effects are not confounded by the effects of sex chromosomes. Results revealed pervasive sex differences in mitochondrial effects, including effects on energetics and aging involving nuclear interactions throughout the genome. Using single-individual RNA sequencing, sex differences were found to explain more than 80% of the variance in gene expression. Males had higher expression of mitochondrial genes and mitochondrially targeted proteins (MTPs) involved in oxidative phosphorylation (OXPHOS), while females had elevated expression of non-OXPHOS MTPs, indicating strongly sex-dimorphic energy metabolism at the whole organism level. Comparison of reciprocal F1 hybrids allowed insights into the nature of mito-nuclear interactions, showing both mitochondrial effects on nuclear expression, and nuclear effects on mitochondrial expression. While based on a small set of crosses, sex-specific increases in mitochondrial expression with age were associated with longer life. Network analyses identified nuclear components of strong mito-nuclear interactions and found them to be sexually dimorphic. These results highlight the profound impact of mitochondria and mito-nuclear interactions on sex- and age-specific gene expression.


Subject(s)
Mitochondria , Sex Chromosomes , Animals , Female , Male , Mitochondria/genetics , Mitochondria/metabolism , Sex Chromosomes/genetics , Aging/genetics , Aging/metabolism , Oxidative Phosphorylation , Sex Characteristics , DNA, Mitochondrial/genetics , Cell Nucleus/metabolism , Cell Nucleus/genetics , Gene Expression Regulation , Energy Metabolism/genetics
10.
Biochemistry (Mosc) ; 89(4): 688-700, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38831505

ABSTRACT

Eukaryotic cells are characterized by a high degree of compartmentalization of their internal contents, which ensures precise and controlled regulation of intracellular processes. During many processes, including different stages of transcription, dynamic membraneless compartments termed biomolecular condensates are formed. Transcription condensates contain various transcription factors and RNA polymerase and are formed by high- and low-specificity interactions between the proteins, DNA, and nearby RNA. This review discusses recent data demonstrating important role of nonspecific multivalent protein-protein and RNA-protein interactions in organization and regulation of transcription.


Subject(s)
Transcription, Genetic , Humans , Transcription Factors/metabolism , DNA-Directed RNA Polymerases/metabolism , DNA/metabolism , DNA/chemistry , RNA/metabolism , RNA/chemistry , Biomolecular Condensates/metabolism , Biomolecular Condensates/chemistry , Animals , Gene Expression Regulation
11.
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
12.
Sci Rep ; 14(1): 12786, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38834626

ABSTRACT

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease marked by inflammatory cell infiltration and joint damage. The Chinese government has approved the prescription medication sinomenine (SIN), an effective anti-inflammation drug, for treating RA. This study evaluated the possible anti-inflammatory actions of SIN in RA based on bioinformatics analysis and experiments. Six microarray datasets were acquired from the gene expression omnibus (GEO) database. We used R software to identify differentially expressed genes (DEGs) and perform function evaluations. The CIBERSORT was used to calculate the abundance of 22 infiltrating immune cells. The weighted gene co-expression network analysis (WGCNA) was used to discover genes associated with M1 macrophages. Four public datasets were used to predict the genes of SIN. Following that, function enrichment analysis for hub genes was performed. The cytoHubba and least absolute shrinkage and selection operator (LASSO) were employed to select hub genes, and their diagnostic effectiveness was predicted using the receiver operator characteristic (ROC) curve. Molecular docking was undertaken to confirm the affinity between the SIN and hub gene. Furthermore, the therapeutic efficacy of SIN was validated in LPS-induced RAW264.7 cells line using Western blot and Enzyme-linked immunosorbent assay (ELISA). The matrix metalloproteinase 9 (MMP9) was identified as the hub M1 macrophages-related biomarker in RA using bioinformatic analysis and molecular docking. Our study indicated that MMP9 took part in IL-17 and TNF signaling pathways. Furthermore, we found that SIN suppresses the MMP9 protein overexpression and pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in the LPS-induced RAW264.7 cell line. In conclusion, our work sheds new light on the pathophysiology of RA and identifies MMP9 as a possible RA key gene. In conclusion, the above findings demonstrate that SIN, from an emerging research perspective, might be a potential cost-effective anti-inflammatory medication for treating RA.


Subject(s)
Arthritis, Rheumatoid , Computational Biology , Cytokines , Matrix Metalloproteinase 9 , Morphinans , Morphinans/pharmacology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Mice , Animals , RAW 264.7 Cells , Computational Biology/methods , Cytokines/metabolism , Humans , Molecular Docking Simulation , Gene Expression Regulation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Anti-Inflammatory Agents/pharmacology
13.
PLoS One ; 19(6): e0304530, 2024.
Article in English | MEDLINE | ID: mdl-38829908

ABSTRACT

Rheumatoid arthritis (RA) is a systemic immune-mediated disease characterized by joint inflammation and destruction. The disease typically affects small joints in the hands and feet, later progressing to involve larger joints such as the knees, shoulders, and hips. While the reasons for these joint-specific differences are unclear, distinct epigenetic patterns associated with joint location have been reported. In this study, we evaluated the unique epigenetic landscapes of fibroblast-like synoviocytes (FLS) from hip and knee synovium in RA patients, focusing on the expression and regulation of Homeobox (HOX) transcription factors. These highly conserved genes play a critical role in embryonic development and are known to maintain distinct expression patterns in various adult tissues. We found that several HOX genes, especially HOXD10, were differentially expressed in knee FLS compared with hip FLS. Epigenetic differences in chromatin accessibility and histone marks were observed in HOXD10 promoter between knee and hip FLS. Histone modification, particularly histone acetylation, was identified as an important regulator of HOXD10 expression. To understand the mechanism of differential HOXD10 expression, we inhibited histone deacetylases (HDACs) with small molecules and siRNA. We found that HDAC1 blockade or deficiency normalized the joint-specific HOXD10 expression patterns. These observations suggest that epigenetic differences, specifically histone acetylation related to increased HDAC1 expression, play a crucial role in joint-specific HOXD10 expression. Understanding these mechanisms could provide insights into the regional aspects of RA and potentially lead to therapeutic strategies targeting specific patterns of joint involvement during the course of disease.


Subject(s)
Arthritis, Rheumatoid , Epigenesis, Genetic , Fibroblasts , Homeodomain Proteins , Synoviocytes , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Synoviocytes/metabolism , Synoviocytes/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Promoter Regions, Genetic , Knee Joint/pathology , Knee Joint/metabolism , Gene Expression Regulation , Histones/metabolism , Acetylation , Hip Joint/pathology , Hip Joint/metabolism
14.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38830772

ABSTRACT

Nucleosome positioning is a key factor for transcriptional regulation. Nucleosomes regulate the dynamic accessibility of chromatin and interact with the transcription machinery at every stage. Influences to steer nucleosome positioning are diverse, and the according importance of the DNA sequence in contrast to active chromatin remodeling has been the subject of long discussion. In this study, we evaluate the functional role of DNA sequence for all major elements along the process of transcription. We developed a random forest classifier based on local DNA structure that assesses the sequence-intrinsic support for nucleosome positioning. On this basis, we created a simple data resource that we applied genome-wide to the human genome. In our comprehensive analysis, we found a special role of DNA in mediating the competition of nucleosomes with cis-regulatory elements, in enabling steady transcription, for positioning of stable nucleosomes in exons, and for repelling nucleosomes during transcription termination. In contrast, we relate these findings to concurrent processes that generate strongly positioned nucleosomes in vivo that are not mediated by sequence, such as energy-dependent remodeling of chromatin.


Subject(s)
Chromatin Assembly and Disassembly , DNA , Gene Expression Regulation , Nucleosomes , Transcription, Genetic , Nucleosomes/metabolism , Nucleosomes/genetics , Humans , Chromatin Assembly and Disassembly/genetics , DNA/genetics , DNA/metabolism , Chromatin/metabolism , Chromatin/genetics , Genome, Human , Base Sequence
15.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38830770

ABSTRACT

Post-transcriptional regulation of immune-related transcripts by RNA-binding proteins (RBPs) impacts immune cell responses, including mast cell functionality. Despite their importance in immune regulation, the functional role of most RBPs remains to be understood. By manipulating the expression of specific RBPs in murine mast cells, coupled with mass spectrometry and transcriptomic analyses, we found that the Regnase family of proteins acts as a potent regulator of mast cell physiology. Specifically, Regnase-1 is required to maintain basic cell proliferation and survival, whereas both Regnase-1 and -3 cooperatively regulate the expression of inflammatory transcripts upon activation, with Tnf being a primary target in both human and mouse cells. Furthermore, Regnase-3 directly interacts with Regnase-1 in mast cells and is necessary to restrain Regnase-1 expression through the destabilization of its transcript. Overall, our study identifies protein interactors of endogenously expressed Regnase factors, characterizes the regulatory interplay between Regnase family members in mast cells, and establishes their role in the control of mast cell homeostasis and inflammatory responses.


Subject(s)
Cell Survival , Cytokines , Mast Cells , Mast Cells/metabolism , Animals , Mice , Humans , Cytokines/metabolism , Cell Survival/genetics , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/genetics , Ribonucleases/metabolism , Ribonucleases/genetics , Gene Expression Regulation , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Mice, Inbred C57BL , Cell Proliferation , Inflammation/metabolism , Transcription Factors
16.
Genome Biol ; 25(1): 142, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38825692

ABSTRACT

BACKGROUND: Like its parent base 5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC) is a direct epigenetic modification of cytosines in the context of CpG dinucleotides. 5hmC is the most abundant oxidized form of 5mC, generated through the action of TET dioxygenases at gene bodies of actively-transcribed genes and at active or lineage-specific enhancers. Although such enrichments are reported for 5hmC, to date, predictive models of gene expression state or putative regulatory regions for genes using 5hmC have not been developed. RESULTS: Here, by using only 5hmC enrichment in genic regions and their vicinity, we develop neural network models that predict gene expression state across 49 cell types. We show that our deep neural network models distinguish high vs low expression state utilizing only 5hmC levels and these predictive models generalize to unseen cell types. Further, in order to leverage 5hmC signal in distal enhancers for expression prediction, we employ an Activity-by-Contact model and also develop a graph convolutional neural network model with both utilizing Hi-C data and 5hmC enrichment to prioritize enhancer-promoter links. These approaches identify known and novel putative enhancers for key genes in multiple immune cell subsets. CONCLUSIONS: Our work highlights the importance of 5hmC in gene regulation through proximal and distal mechanisms and provides a framework to link it to genome function. With the recent advances in 6-letter DNA sequencing by short and long-read techniques, profiling of 5mC and 5hmC may be done routinely in the near future, hence, providing a broad range of applications for the methods developed here.


Subject(s)
5-Methylcytosine , Enhancer Elements, Genetic , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Humans , Neural Networks, Computer , Gene Expression Regulation , Epigenesis, Genetic , DNA Methylation
17.
RNA Biol ; 21(1): 31-44, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38828710

ABSTRACT

Non-thermal plasma, a partially ionized gas, holds significant potential for clinical applications, including wound-healing support, oral therapies, and anti-tumour treatments. While its applications showed promising outcomes, the underlying molecular mechanisms remain incompletely understood. We thus apply non-thermal plasma to mouse auricular skin and conducted non-coding RNA sequencing, as well as single-cell blood sequencing. In a time-series analysis (five timepoints spanning 2 hours), we compare the expression of microRNAs in the plasma-treated left ears to the unexposed right ears of the same mice as well as to the ears of unexposed control mice. Our findings indicate specific effects in the treated ears for a set of five miRNAs: mmu-miR-144-5p, mmu-miR-144-3p, mmu-miR-142a-5p, mmu-miR-223-3p, and mmu-miR-451a. Interestingly, mmu-miR-223-3p also exhibits an increase over time in the right non-treated ear of the exposed mice, suggesting systemic effects. Notably, this miRNA, along with mmu-miR-142a-5p and mmu-miR-144-3p, regulates genes and pathways associated with wound healing and tissue regeneration (namely ErbB, FoxO, Hippo, and PI3K-Akt signalling). This co-regulation is particularly remarkable considering the significant seed dissimilarities among the miRNAs. Finally, single-cell sequencing of PBMCs reveals the downregulation of 12 from 15 target genes in B-cells, Cd4+ and Cd8+ T-cells. Collectively, our data provide evidence for a systemic effect of non-thermal plasma.


Subject(s)
Gene Expression Regulation , MicroRNAs , Plasma Gases , Skin , MicroRNAs/genetics , Animals , Mice , Skin/metabolism , Plasma Gases/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Profiling , Wound Healing/drug effects , Signal Transduction , Immune System/metabolism
18.
J Cell Mol Med ; 28(11): e18447, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38837574

ABSTRACT

The purpose of this study was to identify the mechanisms underlying the involvement of glycolytic genes in pulmonary arterial hypertension (PAH). This study involved downloading 3 datasets from the GEO database at the National Center for Biotechnology Information. The datasets were processed to obtain expression matrices for analysis. Genes involved in glycolysis-related pathways were obtained, and genes related to glycolysis were selected based on significant differences in expression. Gene Ontology functional annotation analysis, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, and GSEA enrichment analysis were performed on the DEGs. Combining LASSO regression with SVM-RFE machine learning technology, a PAH risk prediction model based on glycolysis related gene expression was constructed, and CIBERSORTx technology was used to analyse the immune cell composition of PAH patients. Gene enrichment analysis revealed that the DEGs work synergistically across multiple biological pathways. A total of 6 key glycolysis-related genes were selected using LASSO regression and SVM. A bar plot was constructed to evaluate the weights of the key genes and predict the risk of PAH. The clinical application value and predictive accuracy of the model were assessed. Immunological feature analysis revealed significant correlations between key glycolysis-related genes and the abundances of different immune cell types. The glycolysis genes (ACSS2, ALAS2, ALDH3A1, ADOC3, NT5E, and TALDO1) identified in this study play important roles in the development of pulmonary arterial hypertension, providing new evidence for the involvement of glycolysis in PAH.


Subject(s)
Computational Biology , Glycolysis , Pulmonary Arterial Hypertension , Humans , Glycolysis/genetics , Computational Biology/methods , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Gene Ontology , Gene Expression Regulation , Databases, Genetic
19.
Sci Adv ; 10(23): eadk2693, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38838155

ABSTRACT

T helper 1 (TH1) cell identity is defined by the expression of the lineage-specifying transcription factor T-bet. Here, we examine the influence of T-bet expression heterogeneity on subset plasticity by leveraging cell sorting of distinct in vivo-differentiated TH1 cells based on their quantitative expression of T-bet and interferon-γ. Heterogeneous T-bet expression states were regulated by virus-induced type I interferons and were stably maintained even after secondary viral infection. Exposed to alternative differentiation signals, the sorted subpopulations exhibited graded levels of plasticity, particularly toward the TH2 lineage: T-bet quantities were inversely correlated with the ability to express the TH2 lineage-specifying transcription factor GATA-3 and TH2 cytokines. Reprogramed TH1 cells acquired graded mixed TH1 + TH2 phenotypes with a hybrid epigenetic landscape. Continuous presence of T-bet in differentiated TH1 cells was essential to ensure TH1 cell stability. Thus, innate cytokine signals regulate TH1 cell plasticity via an individual cell-intrinsic rheostat to enable T cell subset adaptation to subsequent challenges.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Plasticity , T-Box Domain Proteins , Th1 Cells , Th2 Cells , Th1 Cells/immunology , Th1 Cells/metabolism , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Animals , Cell Lineage/genetics , Th2 Cells/immunology , Th2 Cells/metabolism , Mice , GATA3 Transcription Factor/metabolism , GATA3 Transcription Factor/genetics , Interferon-gamma/metabolism , Gene Expression Regulation , Cytokines/metabolism
20.
Article in English | MEDLINE | ID: mdl-38847150

ABSTRACT

BACKGROUND AND OBJECTIVE: Nitrate, as nitric oxide (NO) donor, has been suggested as a nutrition-based treatment for decreasing the risk of menopause-related obesity. This study aimed to specify the effects of chronic inorganic nitrate administration on uncoupling protein-1 (UCP-1), peroxisome proliferator-activated-receptor-947; (PPAR-947;) coactivator-1945; (PGC-1945;), and PPAR-947; expression in gonadal adipose tissue (GAT) of ovariectomized (OVX) rats. METHODS: Female rats were assigned to 3 groups: Control, OVX, and OVX+nitrate (n=7/group), which consumed water containing inorganic nitrate (100 mg/L) for 9 months. At month 9, GAT was used for the measurement of NO metabolites (NOx), mRNA levels of NO synthases (endothelial (eNOS), inducible (iNOS), neuronal (nNOS)), and mRNA and protein levels of UCP-1, PGC-1945;, and PPAR-947;. RESULTS: OVX rats had lower NOx concentration (45%) and eNOS (38%) and nNOS (30%) expression in GAT that was restored to normal values following nitrate administration. OVX rats had significantly lower mRNA and protein levels of UCP-1 (83% and 30%), PGC-1945; (65% and 39%), and PPAR-947; (66% and 34.5%) in GAT. Chronic inorganic nitrate administration in OVXrats increased mRNA and protein levels of UCP-1 (128% and 34%), PGC-1945; (115% and 43%), and PPAR-947; (236% and 38%), respectively. CONCLUSION: In OVX rats, chronic nitrate administration increased gene and protein levels of UCP-1, PGC-1945;, and PPAR-947; in GAT, indicating the anti-obesity effects of nitrate are partially mediated by the white adipose tissue (WAT) browning. Moreover, the stimulatory effect of inorganic nitrate on the WAT browning in OVX rats was associated with blunting the OVXinduced NO deficiency in GAT.


Subject(s)
Adipose Tissue, Brown , Nitrates , Ovariectomy , Rats, Wistar , Uncoupling Protein 1 , Animals , Female , Nitrates/administration & dosage , Nitrates/metabolism , Rats , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Nitric Oxide/metabolism , Gene Expression Regulation/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
SELECTION OF CITATIONS
SEARCH DETAIL
...