Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.994
Filtrar
1.
Methods Mol Biol ; 2856: 327-339, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39283462

RESUMEN

Disentangling the relationship of enhancers and genes is an ongoing challenge in epigenomics. We present STARE, our software to quantify the strength of enhancer-gene interactions based on enhancer activity and chromatin contact data. It implements the generalized Activity-by-Contact (gABC) score, which allows predicting putative target genes of candidate enhancers over any desired genomic distance. The only requirement for its application is a measurement of enhancer activity. In addition to regulatory interactions, STARE calculates transcription factor (TF) affinities on gene level. We illustrate its usage on a public single-cell data set of the human heart by predicting regulatory interactions on cell type level, by giving examples on how to integrate them with other data modalities, and by constructing TF affinity matrices.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Epigenómica , Programas Informáticos , Humanos , Cromatina/genética , Cromatina/metabolismo , Epigenómica/métodos , Epigenoma , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Biología Computacional/métodos
2.
Methods Mol Biol ; 2856: 341-356, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39283463

RESUMEN

To reveal gene regulation mechanisms, it is essential to understand the role of regulatory elements, which are possibly distant from gene promoters. Integrative analysis of epigenetic and transcriptomic data can be used to gain insights into gene-expression regulation in specific phenotypes. Here, we discuss STITCHIT, an approach to dissect epigenetic variation in a gene-specific manner across many samples for the identification of regulatory elements without relying on peak calling algorithms. The obtained genomic regions are then further refined using a regularized linear model approach, which can also be used to predict gene expression. We illustrate the use of STITCHIT using H3k27ac ChIP-seq and RNA-seq data from the International Human Epigenome Consortium (IHEC).


Asunto(s)
Epigénesis Genética , Epigenómica , Transcriptoma , Humanos , Epigenómica/métodos , Transcriptoma/genética , Elementos de Facilitación Genéticos , Programas Informáticos , Biología Computacional/métodos , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Regulación de la Expresión Génica , Algoritmos , Histonas/genética , Histonas/metabolismo , Perfilación de la Expresión Génica/métodos
3.
Methods Mol Biol ; 2856: 357-400, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39283464

RESUMEN

Three-dimensional (3D) chromatin interactions, such as enhancer-promoter interactions (EPIs), loops, topologically associating domains (TADs), and A/B compartments, play critical roles in a wide range of cellular processes by regulating gene expression. Recent development of chromatin conformation capture technologies has enabled genome-wide profiling of various 3D structures, even with single cells. However, current catalogs of 3D structures remain incomplete and unreliable due to differences in technology, tools, and low data resolution. Machine learning methods have emerged as an alternative to obtain missing 3D interactions and/or improve resolution. Such methods frequently use genome annotation data (ChIP-seq, DNAse-seq, etc.), DNA sequencing information (k-mers and transcription factor binding site (TFBS) motifs), and other genomic properties to learn the associations between genomic features and chromatin interactions. In this review, we discuss computational tools for predicting three types of 3D interactions (EPIs, chromatin interactions, and TAD boundaries) and analyze their pros and cons. We also point out obstacles to the computational prediction of 3D interactions and suggest future research directions.


Asunto(s)
Cromatina , Aprendizaje Profundo , Cromatina/genética , Cromatina/metabolismo , Humanos , Biología Computacional/métodos , Aprendizaje Automático , Genómica/métodos , Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Sitios de Unión , Genoma , Programas Informáticos
4.
J Ethnopharmacol ; 336: 118706, 2025 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-39186989

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Ganoderma lucidum (G. lucidum) has been widely used as adjuvant of anti-tumor therapy for variety tumors. The bioactive ingredients of G. lucidum mainly include triterpenes, such as Ganoderic acid A, Ganoderic acid B, Ganoderenic acid A, Ganoderenic acid B, Ganoderenic acid D, and Ganoderic acid X. However, the effects and underlying mechanisms of G. lucidum are often challenging in hepatocellular carcinoma (HCC) treatment. AIM OF THE STUDY: To explore the potential role and mechanism of enhancer-associated lncRNAs (en-lncRNAs) in G. lucidum treated HCC through the in vivo and in vitro experiments. MATERIALS AND METHODS: Hepa1-6-bearing C57 BL/6 mice model were established to evaluate the therapeutic efficacy of G. lucidum treated HCC. Ki67 and TUNEL staining were used to detect the tumor cell proliferation and apoptosis in vivo. The Mouse lncRNA 4*180K array was implemented to identify the differentially expressed (DE) lncRNAs and mRNAs of G. lucidum treated tumor mice. The constructed lncRNA-mRNA co-expression network and bioinformatics analysis were used to selected core en-lncRNAs and its neighboring genes. The UPLC-MS method was used to identify the triterpenes of G. lucidum, and the in vitro experiments were used to verify which triterpene monomers regulated en-lncRNAs in tumor cells. Finally, a stable knockdown/overexpression cell lines were used to confirm the relationship between en-lncRNA and neighboring gene. RESULTS: Ki67 and TUNEL staining demonstrated G. lucidum significantly inhibited tumor growth, suppressed cell proliferation and induced apoptosis in vivo. Transcriptomic analysis revealed the existence of 126 DE lncRNAs high correlated with 454 co-expressed mRNAs in G. lucidum treated tumor mice. Based on lncRNA-mRNA network and qRT-PCR validation, 6 core lncRNAs were selected and considered high correlated with G. lucidum treatment. Bioinformatics analysis revealed FR036820 and FR121302 might act as enhancers, and qRT-PCR results suggested FR121302 might enhance Popdc2 mRNA level in HCC. Furthermore, 6 main triterpene monomers of G. lucidum were identified by UPLC-MS method, and in vitro experiments showed FR121302 and Popdc2 were significantly suppressed by Ganoderenic acid A and Ganoderenic acid B, respectively. The knock/overexpression results demonstrated that FR121302 activating and enhancing Popdc2 expression levels, and Ganoderenic acid A and Ganoderenic acid B dramatically suppressed FR121302 and decreased Popdc2 level in Hepa1-6 cells. CONCLUSIONS: Enhancer-associated lncRNA plays a crucial role as an enhancer during hepatocarcinogenesis, and triterpenes of G. lucidum significantly inhibited tumor cell proliferation and induced apoptosis by regulating en-lncRNAs. Our study demonstrated Ganoderenic acid A and Ganoderenic acid B suppressed en-lncRNA FR121302 may be one of the critical strategies of G. lucidum inhibit hepatocellular carcinoma growth.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular , Proliferación Celular , Neoplasias Hepáticas , Ratones Endogámicos C57BL , ARN Largo no Codificante , Reishi , Triterpenos , Animales , Triterpenos/farmacología , Triterpenos/aislamiento & purificación , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Reishi/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ratones , Línea Celular Tumoral , Masculino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/aislamiento & purificación
5.
Biochim Biophys Acta Gene Regul Mech ; 1867(4): 195059, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39226990

RESUMEN

Over the past decade, regulatory non-coding RNAs (ncRNAs) produced by RNA Pol II have been revealed as meaningful players in various essential cellular functions. In particular, thousands of ncRNAs are produced at transcriptional regulatory elements such as enhancers and promoters, where they may exert multiple functions to regulate proper development, cellular programming, transcription or genomic stability. Here, we review the mechanisms involving these regulatory element-associated ncRNAs, and particularly enhancer RNAs (eRNAs) and PROMoter uPstream Transcripts (PROMPTs). We contextualize the mechanisms described to the processing and degradation of these short lived RNAs. We summarize recent findings explaining how ncRNAs operate locally at promoters and enhancers, or further away, either shortly after their production by RNA Pol II, or through post-transcriptional stabilization. Such discoveries lead to a converging model accounting for how ncRNAs influence cellular fate, by acting on transcription and chromatin structure, which may further involve factors participating to 3D nuclear organization.

6.
Development ; 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289870

RESUMEN

Understanding how cell identity is encoded by the genome and acquired during differentiation is a central challenge in cell biology. We have developed a theoretical framework called EnhancerNet, which models the regulation of cell identity through the lens of transcription factor (TF)-enhancer interactions. We demonstrate that autoregulation in these interactions imposes a constraint on the model, resulting in simplified dynamics that can be parameterized from observed cell identities. Despite its simplicity, EnhancerNet recapitulates a broad range of experimental observations on cell identity dynamics, including enhancer selection, cell fate induction, hierarchical differentiation through multipotent progenitor states, and direct reprogramming by TF overexpression. The model makes specific quantitative predictions, reproducing known reprogramming recipes and the complex hematopoietic differentiation hierarchy without fitting unobserved parameters. EnhancerNet provides insights into how new cell types could evolve and highlights the functional importance of distal regulatory elements with dynamic chromatin in multicellular evolution.

7.
Genome Med ; 16(1): 112, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39272130

RESUMEN

BACKGROUND: X-linked acrogigantism (X-LAG; MIM: 300942) is a severe form of pituitary gigantism caused by chromosome Xq26.3 duplications involving GPR101. X-LAG-associated duplications disrupt the integrity of the topologically associating domain (TAD) containing GPR101 and lead to the formation of a neo-TAD that drives pituitary GPR101 misexpression and gigantism. As X-LAG is fully penetrant and heritable, duplications involving GPR101 identified on prenatal screening studies, like amniocentesis, can pose an interpretation challenge for medical geneticists and raise important concerns for patients and families. Therefore, providing robust information on the functional genomic impact of such duplications has important research and clinical value with respect to gene regulation and triplosensitivity traits. METHODS: We employed 4C/HiC-seq as a clinical tool to determine the functional impact of incidentally discovered GPR101 duplications on TAD integrity in three families. After defining duplications and breakpoints around GPR101 by clinical-grade and high-density aCGH, we constructed 4C/HiC chromatin contact maps for our study population and compared them with normal and active (X-LAG) controls. RESULTS: We showed that duplications involving GPR101 that preserved the centromeric invariant TAD boundary did not generate a pathogenic neo-TAD and that ectopic enhancers were not adopted. This allowed us to discount presumptive/suspected X-LAG diagnoses and GPR101 misexpression, obviating the need for intensive clinical follow-up. CONCLUSIONS: This study highlights the importance of TAD boundaries and chromatin interactions in determining the functional impact of copy number variants and provides proof-of-concept for using 4C/HiC-seq as a clinical tool to acquire crucial information for genetic counseling and to support clinical decision-making in cases of suspected TADopathies.


Asunto(s)
Cromatina , Receptores Acoplados a Proteínas G , Humanos , Receptores Acoplados a Proteínas G/genética , Cromatina/genética , Cromatina/metabolismo , Femenino , Masculino , Duplicación de Gen , Duplicación Cromosómica , Cromosomas Humanos X/genética , Linaje
8.
Adv Exp Med Biol ; 1460: 329-356, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39287857

RESUMEN

Obese subjects exhibit lower adipose tissue oxygen consumption in accordance with the lower adipose tissue blood flow. Thereby, compared to lean subjects, obese individuals have almost half lower capillary density and more than half lower vascular endothelial growth factor (VEGF). The VEGF expression together with hypoxia-inducible transcription factor-1 alpha (HIF-1α) activity also requires phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR)-mediated signaling. Especially HIF-1α is an important signaling molecule for hypoxia to induce the inflammatory responses. Hypoxia contributes to several biological functions, such as angiogenesis, cell proliferation, apoptosis, inflammation, and insulin resistance (IR). Pathogenesis of obesity-related comorbidities is attributed to intermittent hypoxia (IH), which is mostly observed in visceral obesity. Proinflammatory phenotype of the adipose tissue is a crucial link between IH and the development of IR. Inhibition of adaptive unfolded protein response (UPR) in hypoxia increases ß cell death. Moreover, deletion of HIF-1α worsens ß cell function. Oxidative stress, as well as the release of proinflammatory cytokines/adipokines in obesity, is proportional to the severity of IH. Reactive oxygen species (ROS) generation at mitochondria is responsible for propagation of the hypoxic signal; however, mitochondrial ROS production is required for hypoxic HIF-1α protein stabilization. Alterations in oxygen availability of adipose tissue directly affect the macrophage polarization and are responsible for the dysregulated adipocytokines production in obesity. Hypoxia both inhibits adipocyte differentiation from preadipocytes and macrophage migration from the hypoxic adipose tissue. Upon reaching a hypertrophic threshold beyond the adipocyte fat loading capacity, excess extracellular matrix (ECM) components are deposited, causing fibrosis. HIF-1α initiates the whole pathological process of fibrosis and inflammation in the obese adipose tissue. In addition to stressed adipocytes, hypoxia contributes to immune cell migration and activation which further aggravates adipose tissue fibrosis. Therefore, targeting HIF-1α might be an efficient way to suppress hypoxia-induced pathological changes in the ECM. The fibrosis score of adipose tissue correlates negatively with the body mass index and metabolic parameters. Inducers of browning/beiging adipocytes and adipokines, as well as modulations of matrix remodeling enzyme inhibitors, and associated gene regulators, are potential pharmacological targets for treating obesity.


Asunto(s)
Tejido Adiposo , Obesidad , Humanos , Obesidad/metabolismo , Obesidad/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Animales , Hipoxia/metabolismo , Transducción de Señal , Resistencia a la Insulina
9.
Adv Exp Med Biol ; 1460: 595-627, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39287866

RESUMEN

In obesity, the process of adipogenesis largely determines the number of adipocytes in body fat depots. Adipogenesis is regulated by several adipocyte-selective micro-ribonucleic acids (miRNAs) and transcription factors that modulate adipocyte proliferation and differentiation. However, some miRNAs block the expression of master regulators of adipogenesis. Since the specific miRNAs display different expressions during adipogenesis, in mature adipocytes and permanent obesity, their use as biomarkers or therapeutic targets is feasible. Upregulated miRNAs in persistent obesity are downregulated during adipogenesis. Moreover, some of the downregulated miRNAs in obese individuals are upregulated in mature adipocytes. Induction of adipocyte stress and hypertrophy leads to the release of adipocyte-derived exosomes (AdEXs) that contain the cargo molecules, miRNAs. miRNAs are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. While each miRNA targets multiple messenger RNAs (mRNAs), which may coordinate or antagonize each other's functions, several miRNAs are dysregulated in other tissues during obesity-related comorbidities. Deletion of the miRNA-processing enzyme DICER in pro-opiomelanocortin-expressing cells results in obesity, which is characterized by hyperphagia, increased adiposity, hyperleptinemia, defective glucose metabolism, and alterations in the pituitary-adrenal axis. In recent years, RNA-based therapeutical approaches have entered clinical trials as novel therapies against overweight and its complications. Development of lipid droplets, macrophage accumulation, macrophage polarization, tumor necrosis factor receptor-associated factor 6 activity, lipolysis, lipotoxicity, and insulin resistance are effectively controlled by miRNAs. Thereby, miRNAs as epigenetic regulators are used to determine the new gene transcripts and therapeutic targets.


Asunto(s)
Adipogénesis , Epigénesis Genética , MicroARNs , Obesidad , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Obesidad/genética , Obesidad/metabolismo , Adipogénesis/genética , Animales , Adipocitos/metabolismo , Exosomas/metabolismo , Exosomas/genética , Regulación de la Expresión Génica
10.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39273113

RESUMEN

Sodium tungstate (Na2WO4) normalizes glucose metabolism in the liver and muscle, activating the Mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway. Because this pathway controls neuronal survival and differentiation, we investigated the effects of Na2WO4 in mouse Neuro2a and human SH-SY5Y neuroblastoma monolayer cell cultures. Na2WO4 promotes differentiation to cholinergic neurites via an increased G1/G0 cell cycle in response to the synergic activation of the Phosphatidylinositol 3-kinase (PI3K/Akt) and ERK1/2 signaling pathways. In Neuro2a cells, Na2WO4 increases protein synthesis by activating the mechanistic target of rapamycin (mTOR) and S6K kinases and GLUT3-mediated glucose uptake, providing the energy and protein synthesis needed for neurite outgrowth. Furthermore, Na2WO4 increased the expression of myocyte enhancer factor 2D (MEF2D), a member of a family of transcription factors involved in neuronal survival and plasticity, through a post-translational mechanism that increases its half-life. Site-directed mutations of residues involved in the sumoylation of the protein abrogated the positive effects of Na2WO4 on the MEF2D-dependent transcriptional activity. In addition, the neuroprotective effects of Na2WO4 were evaluated in the presence of advanced glycation end products (AGEs). AGEs diminished neurite differentiation owing to a reduction in the G1/G0 cell cycle, concomitant with lower expression of MEF2D and the GLUT3 transporter. These negative effects were corrected in both cell lines after incubation with Na2WO4. These findings support the role of Na2WO4 in neuronal plasticity, albeit further experiments using 3D cultures, and animal models will be needed to validate the therapeutic potential of the compound.


Asunto(s)
Proyección Neuronal , Fármacos Neuroprotectores , Compuestos de Tungsteno , Humanos , Proyección Neuronal/efectos de los fármacos , Animales , Línea Celular Tumoral , Compuestos de Tungsteno/farmacología , Ratones , Fármacos Neuroprotectores/farmacología , Neuroprotección/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Neuritas/metabolismo , Neuritas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos
11.
bioRxiv ; 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39229027

RESUMEN

Identifying cell type-specific enhancers in the brain is critical to building genetic tools for investigating the mammalian brain. Computational methods for functional enhancer prediction have been proposed and validated in the fruit fly and not yet the mammalian brain. We organized the 'Brain Initiative Cell Census Network (BICCN) Challenge: Predicting Functional Cell Type-Specific Enhancers from Cross-Species Multi-Omics' to assess machine learning and feature-based methods designed to nominate enhancer DNA sequences to target cell types in the mouse cortex. Methods were evaluated based on in vivo validation data from hundreds of cortical cell type-specific enhancers that were previously packaged into individual AAV vectors and retro-orbitally injected into mice. We find that open chromatin was a key predictor of functional enhancers, and sequence models improved prediction of non-functional enhancers that can be deprioritized as opposed to pursued for in vivo testing. Sequence models also identified cell type-specific transcription factor codes that can guide designs of in silico enhancers. This community challenge establishes a benchmark for enhancer prioritization algorithms and reveals computational approaches and molecular information that are crucial for the identification of functional enhancers for mammalian cortical cell types. The results of this challenge bring us closer to understanding the complex gene regulatory landscape of the mammalian brain and help us design more efficient genetic tools and potential gene therapies for human neurological diseases.

12.
bioRxiv ; 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39257788

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer without effective treatments. It is characterized by activating KRAS mutations and p53 alterations. However, how these mutations dysregulate cancer-cell-intrinsic gene programs to influence the immune landscape of the tumor microenvironment (TME) remains poorly understood. Here, we show that p53R172H establishes an immunosuppressive TME, diminishes the efficacy of immune checkpoint inhibitors (ICIs), and enhances tumor growth. Our findings reveal that the upregulation of the immunosuppressive chemokine Cxcl1 mediates these pro-tumorigenic functions of p53R172H. Mechanistically, we show that p53R172H associates with the distal enhancers of the Cxcl1 gene, increasing enhancer activity and Cxcl1 expression. p53R172H occupies these enhancers in an NF-κB-pathway-dependent manner, suggesting NF-κB's role in recruiting p53R172H to the Cxcl1 enhancers. Our work uncovers how a common mutation in a tumor-suppressor transcription factor appropriates enhancers, stimulating chemokine expression and establishing an immunosuppressive TME that diminishes ICI efficacy in PDAC.

13.
Cancer Biomark ; 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39269825

RESUMEN

OBJECTIVE: To investigate the impact and potential mechanisms of serum extracellular nano-vesicles (sEVs) miR-412-3p released from sub-centimeter lung nodules with a diameter of ⩽ 10 mm on the malignant biological function of micro-nodular lung cancer (mnLC). METHODS: A total of 87 participants were included and divided into a mnLC group (n= 30), a benign lung nodule (BLN) group (n= 27), and a healthy people control group (n= 30). Transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and Western blot (WB) were used to measure the morphological characteristics and surface markers of sEVs. In vitro analysis, real-time quantitative polymerase chain reaction (RT-qPCR), CCK-8 cell proliferation assay, clone formation assay, Transwell, stem cell sphere-forming assay, and WB assay were conducted to verify the effect of miR-412-3p/TEAD1 signaling axis on the biological function of lung cancer cells through, respectively. Further validation was conducted using the serum sEVs of the participants. RESULTS: The expression level of sEVs-miR-412-3p in the mnLC group was significantly higher than that in the BLN and healthy groups (P< 0.01). In lung cancer cell lines, miR-412-3p can negatively regulate the targeted gene TEAD1. The miR-412-3p/TEAD1 signaling axis is involved in promoting the EMT signaling pathway and regulating the malignant biological functions of lung cancer cell proliferation, migration, and stemness (P< 0.05). In addition, sEVs in the mnLC group significantly promoted lung cancer cell proliferation, migration, and stemness compared to the BLN and healthy groups, inhibited the expression of E-cadherin and TEAD1 in lung cancer cells, and promoted the expression of N-cadherin and Vimentin (P< 0.05). CONCLUSION: sEVs-miR-412-3p could promote the biological process of EMT, and lead to the occurrence of malignant biological behavior in sub-centimeter lung nodules. This provides evidence for the miR-412-3p/TEAD1 signaling axis as a potential therapeutic target for mnLC.

14.
Adv Immunol ; 163: 51-96, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39271259

RESUMEN

Interferon regulatory factor-8 (IRF8) is the lineage determining transcription factor for the type one classical dendritic cell (cDC1) subset, a terminal selector for plasmacytoid dendritic cells and important for the function of monocytes. Studies of Irf8 gene regulation have identified several enhancers controlling its activity during development of progenitors in the bone marrow that precisely regulate expression at distinct developmental stages. Each enhancer responds to distinct transcription factors that are expressed at each stage. IRF8 is first expressed in early progenitors that form the monocyte dendritic cell progenitor (MDP) in response to induction of the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) acting at the Irf8 +56 kb enhancer. IRF8 levels increase further as the MDP transits into the common dendritic cell progenitor (CDP) in response to E protein activity at the Irf8 +41 kb enhancer. Upon Nfil3-induction in CDPs leading to specification of the cDC1 progenitor, abrupt induction of BATF3 forms the JUN/BATF3/IRF8 heterotrimer that activates the Irf8 +32 kb enhancer that sustains Irf8 autoactivation throughout the cDC1 lifetime. Deletions of each of these enhancers has revealed their stage dependent activation. Surprisingly, studies of compound heterozygotes for each combination of enhancer deletions revealed that activation of each subsequent enhancer requires the successful activation of the previous enhancer in strictly cis-dependent mechanism. Successful progression of enhancer activation is finely tuned to alter the functional accessibility of subsequent enhancers to factors active in the next stage of development. The molecular basis for these phenomenon is still obscure but could have implications for genomic regulation in a broader developmental context.


Asunto(s)
Células Dendríticas , Elementos de Facilitación Genéticos , Factores Reguladores del Interferón , Humanos , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Elementos de Facilitación Genéticos/genética , Diferenciación Celular , Regulación de la Expresión Génica , Súper Potenciadores
15.
Yi Chuan ; 46(9): 677-689, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39275868

RESUMEN

The sex determination in mammals refers to the development of an initial bipotential organ, termed the bipotential gonad/genital ridge, into either a testis or an ovary at the early stages of embryonic development, under the precise regulation of transcription factors. SOX9 (SRY-box transcription factor 9) is a multifunctional transcription factor in mammalian development and plays a critical role in sex determination and subsequent male reproductive organs development. Recent studies have shown that several enhancers upstream of SOX9 also play an important role in the process of sex determination. In this review, we summarize the progress on the role of SOX9 and its gonadal enhancers in sex determination. This review will facilitate to understand the regulatory mechanism of sex determination of SOX9 and provides a theoretical basis for the further development of animal sex manipulation technologies.


Asunto(s)
Mamíferos , Factor de Transcripción SOX9 , Procesos de Determinación del Sexo , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Animales , Procesos de Determinación del Sexo/genética , Humanos , Mamíferos/genética , Masculino , Femenino , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica
16.
Biosens Bioelectron ; 266: 116721, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39226753

RESUMEN

Live-cell label-free imaging of a microscopic biological barrier, generally referred to as 'tight junction', was realized by a recently developed electric-double-layer modulation imaging (EDLMI). The method allowed quantitative imaging of barrier integrity in real time, thus being an upper compatible of transepithelial electrical resistance (TEER) which is a conventional standard technique to evaluate spatially averaged barrier integrity. We demonstrate that the quantitative and real-time imaging capability of EDLMI unveils fundamental dynamics of biological barrier, some of which are totally different from conventional understandings.


Asunto(s)
Técnicas Biosensibles , Humanos , Técnicas Biosensibles/métodos , Uniones Estrechas/metabolismo , Impedancia Eléctrica
17.
Int J Pharm ; 665: 124671, 2024 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-39245088

RESUMEN

The study aimed to improve the transdermal permeation of IBU utilizing menthosomes as a vesicular carrier. IBU-loaded menthosomes were formulated by thin film hydration & optimized using 23 factorial designs (Design Expert® version 13 software). In vitro & ex vivo skin permeation analysis of IBU-encapsulated menthosomes was studied across the rat skin sample. In vivo pharmacodynamic activity was studied in an arthritis rat model. The optimized IBU-loaded menthosomes exhibited an optimum vesicle size of 214.2 ± 2.96 nm, Zeta potential of -21.1 ± 2.72 mV, (PDI) Polydispersity Index of 0.267 ± 0.018 with Entrapment efficiency (EE%) of 78.7 ± 2.73 %. The in vitro & ex vivo skin penetration study displayed enhanced release of drug of 77.02 ± 1.0 % and 40.91 ± 0.81 % respectively, compared to conventional liposomes. In vivo pharmacodynamic study on carrageenan-induced paw edema in Wistar albino rats demonstrated superior anti-inflammatory activity of the optimized IBU-encapsulated menthosomes (**p < 0.01) and effective inhibition of paw edema (34.04 ± 0.155 %). The formalin test indicated a significant analgesic effect of optimized formulation during the chronic phase of analgesia (*p < 0.05) compared to the control group. Thus, the developed and optimized drug-loaded menthosomes could serve as a suitable vesicular delivery carrier in enhancing the transdermal delivery of other NSAID drugs.

18.
Brief Bioinform ; 25(5)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39222061

RESUMEN

Harnessing the power of single-cell genomics technologies, single-cell Hi-C (scHi-C) and its derived technologies provide powerful tools to measure spatial proximity between regulatory elements and their target genes in individual cells. Using a global background model, we propose SnapHiC-G, a computational method, to identify long-range enhancer-promoter interactions from scHi-C data. We applied SnapHiC-G to scHi-C datasets generated from mouse embryonic stem cells and human brain cortical cells. SnapHiC-G achieved high sensitivity in identifying long-range enhancer-promoter interactions. Moreover, SnapHiC-G can identify putative target genes for noncoding genome-wide association study (GWAS) variants, and the genetic heritability of neuropsychiatric diseases is enriched for single-nucleotide polymorphisms (SNPs) within SnapHiC-G-identified interactions in a cell-type-specific manner. In sum, SnapHiC-G is a powerful tool for characterizing cell-type-specific enhancer-promoter interactions from complex tissues and can facilitate the discovery of chromatin interactions important for gene regulation in biologically relevant cell types.


Asunto(s)
Elementos de Facilitación Genéticos , Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Análisis de la Célula Individual , Animales , Humanos , Ratones , Análisis de la Célula Individual/métodos , Estudio de Asociación del Genoma Completo/métodos , Genómica/métodos , Biología Computacional/métodos
19.
Cancer Inform ; 23: 11769351241275877, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39238655

RESUMEN

Background: Ovarian cancer has brought serious threats to female health. CCAAT/enhancer binding proteins (C/EBPs) are key transcription factors involved in ovarian cancer. Therefore, comprehensive profiling C/EBPs in ovarian cancer is needed. Methods: A comprehensive analysis concerning C/EBPs in ovarian cancer was performed. Firstly, detailed expression of C/EBP family members was integrally retrieved and then confirmed using immunohistochemistry. The regulatory effects and transcription regulatory functions of C/EBPs were studied by using regulatory network analysis and enrichment analysis. Using survival analysis, receiver operating characteristic curve analysis, and target-disease association analysis, the predictive prognostic value of C/EBPs on survival and drug responsiveness was systematically evaluated. The effects of C/EBPs on tumor immune infiltration were also assessed. Results: Ovarian cancer tissues expressed increased CEBPA, CEBPB, and CEBPG but decreased CEBPD when compared with normal control tissues. The overall alteration frequency of C/EBPs in ovarian cancer was approaching 30%. C/EBP family members formed a reciprocal regulatory network involving carcinogenesis and had pivotal transcription regulatory functions. C/EBPs could affect survival of ovarian cancer and correlated with poor survival outcomes (OS: HR = 1.40, P = .0053 and PFS: HR = 1.41, P = .0036). Besides, expression of CEBPA, CEBPB, CEBPD, and CEBPE could predict platinum and taxane responsiveness of ovarian cancer. C/EBPs also affected immune infiltration of ovarian cancer. Conclusions: C/EBPs were closely involved in ovarian cancer and exerted multiple biological functions. C/EBPs could be exploited as prognostic and predictive biomarkers in ovarian cancer.

20.
Front Nutr ; 11: 1436113, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224182

RESUMEN

Introduction: One major challenge of reducing salt content in food is the risk of the overall taste becoming bland. Enhancing saltiness is an effective strategy for salt reduction, and the development of salt-reduced foods using these saltiness-enhancing flavorants as food additives is underway. However, an increasing number of consumers demand a reduction in additives in clean-label foods. Objective: Enzyme processing of food is an attractive strategy for developing clean-label foods because enzymes are not considered additives. We aimed to improve the saltiness and umami intensity of vegetable soups by enzyme treatment while meeting clean-label requirements. We first optimized the enzymatic reaction conditions of a protease and glutaminase blend and then investigated the synergistic effects of this enzyme blend on the taste of vegetable soup. Results: Sensory evaluations indicated that the reaction products (e.g., protein hydrolysates or amino acids) could enhance the umami, kokumi, and saltiness intensity of vegetable soup supplemented with 0.5% NaCl. Notably, the saltiness intensity ratio of the enzyme-treated soup with 0.50, 0.45, and 0.40% NaCl were increased by 1.31-, 1.16-, and 0.99-fold, respectively, when this ratio for the control soup with 0.50% NaCl was set to 1.0. This indicates a 20% salt reduction rate can be achieved by enzyme treatment. Moreover, we found that these enhancements were synergically caused by enzyme-produced glutamic acid and branched-chain amino acids. Conclusion: Our findings suggest that using enzyme blends of bacterial and fungal proteases and glutaminase is an effective approach to enhancing the saltiness levels of vegetable soups while meeting clean-label requirements.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA