Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Circulation ; 149(19): 1501-1515, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38223978

RESUMEN

BACKGROUND: During the neonatal stage, the cardiomyocyte undergoes a constellation of molecular, cytoarchitectural, and functional changes known collectively as cardiomyocyte maturation to increase myocardial contractility and cardiac output. Despite the importance of cardiomyocyte maturation, the molecular mechanisms governing this critical process remain largely unexplored. METHODS: We leveraged an in vivo mosaic knockout system to characterize the role of Carm1, the founding member of protein arginine methyltransferase, in cardiomyocyte maturation. Using a battery of assays, including immunohistochemistry, immuno-electron microscopy imaging, and action potential recording, we assessed the effect of loss of Carm1 function on cardiomyocyte cell growth, myofibril expansion, T-tubule formation, and electrophysiological maturation. Genome-wide transcriptome profiling, H3R17me2a chromatin immunoprecipitation followed by sequencing, and assay for transposase-accessible chromatin with high-throughput sequencing were used to investigate the mechanisms by which CARM1 (coactivator-associated arginine methyltransferase 1) regulates cardiomyocyte maturation. Finally, we interrogated the human syntenic region to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks for single-nucleotide polymorphisms associated with human heart diseases. RESULTS: We report that mosaic ablation of Carm1 disrupts multiple aspects of cardiomyocyte maturation cell autonomously, leading to reduced cardiomyocyte size and sarcomere thickness, severe loss and disorganization of T tubules, and compromised electrophysiological maturation. Genomics study demonstrates that CARM1 directly activates genes that underlie cardiomyocyte cytoarchitectural and electrophysiological maturation. Moreover, our study reveals significant enrichment of human heart disease-associated single-nucleotide polymorphisms in the human genomic region syntenic to the H3R17me2a chromatin immunoprecipitation followed by sequencing peaks. CONCLUSIONS: This study establishes a critical and multifaceted role for CARM1 in regulating cardiomyocyte maturation and demonstrates that deregulation of CARM1-dependent cardiomyocyte maturation gene expression may contribute to human heart diseases.


Asunto(s)
Epigénesis Genética , Miocitos Cardíacos , Proteína-Arginina N-Metiltransferasas , Animales , Humanos , Ratones , Diferenciación Celular/genética , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo
2.
IEEE Trans Pattern Anal Mach Intell ; 46(5): 3707-3721, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38163314

RESUMEN

Appearance-based gaze estimation has garnered increasing attention in recent years. However, deep learning-based gaze estimation models still suffer from suboptimal performance when deployed in new domains, e.g., unseen environments or individuals. In our previous work, we took this challenge for the first time by introducing a plug-and-play method (PnP-GA) to adapt the gaze estimation model to new domains. The core concept of PnP-GA is to leverage the diversity brought by a group of model variants to enhance the adaptability to diverse environments. In this article, we propose the PnP-GA+ by extending our approach to explore the impact of assembling model variants using three additional perspectives: color space, data augmentation, and model structure. Moreover, we propose an intra-group attention module that dynamically optimizes pseudo-labeling during adaptation. Experimental results demonstrate that by directly plugging several existing gaze estimation networks into the PnP-GA+ framework, it outperforms state-of-the-art domain adaptation approaches on four standard gaze domain adaptation tasks on public datasets. Our method consistently enhances cross-domain performance, and its versatility is improved through various ways of assembling the model group.

3.
Artículo en Inglés | MEDLINE | ID: mdl-38662567

RESUMEN

Human gaze provides valuable information on human focus and intentions, making it a crucial area of research. Recently, deep learning has revolutionized appearance-based gaze estimation. However, due to the unique features of gaze estimation research, such as the unfair comparison between 2D gaze positions and 3D gaze vectors and the different pre-processing and post-processing methods, there is a lack of a definitive guideline for developing deep learning-based gaze estimation algorithms. In this paper, we present a systematic review of the appearance-based gaze estimation methods using deep learning. Firstly, we survey the existing gaze estimation algorithms along the typical gaze estimation pipeline: deep feature extraction, deep learning model design, personal calibration and platforms. Secondly, to fairly compare the performance of different approaches, we summarize the data pre-processing and post-processing methods, including face/eye detection, data rectification, 2D/3D gaze conversion and gaze origin conversion. Finally, we set up a comprehensive benchmark for deep learning-based gaze estimation. We characterize all the public datasets and provide the source code of typical gaze estimation algorithms. This paper serves not only as a reference to develop deep learning-based gaze estimation methods, but also a guideline for future gaze estimation research. The project web page can be found at https://phi-ai.buaa.edu.cn/Gazehub/.

4.
Sci Adv ; 10(8): eadk4694, 2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38381829

RESUMEN

Cardiac regeneration requires coordinated participation of multiple cell types whereby their communications result in transient activation of proregenerative cell states. Although the molecular characteristics and lineage origins of these activated cell states and their contribution to cardiac regeneration have been studied, the extracellular signaling and the intrinsic genetic program underlying the activation of the transient functional cell states remain largely unexplored. In this study, we delineated the chromatin landscapes of the noncardiomyocytes (nonCMs) of the regenerating heart at the single-cell level and inferred the cis-regulatory architectures and trans-acting factors that control cell type-specific gene expression programs. Moreover, further motif analysis and cell-specific genetic manipulations suggest that the macrophage-derived inflammatory signal tumor necrosis factor-α, acting via its downstream transcription factor complex activator protein-1, functions cooperatively with discrete transcription regulators to activate respective nonCM cell types critical for cardiac regeneration. Thus, our study defines the regulatory architectures and intercellular communication principles in zebrafish heart regeneration.


Asunto(s)
Cromatina , Pez Cebra , Animales , Cromatina/genética , Pez Cebra/genética , Regulación del Desarrollo de la Expresión Génica , Corazón/fisiología , Regeneración/genética
5.
Phytomedicine ; 129: 155722, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38733905

RESUMEN

BACKGROUND: Autoimmune hepatitis (AIH), primarily mediated by T cells, is characterized by liver inflammation. Despite the advancements in understanding its pathogenesis, effective therapeutic options are limited. Naringin, a flavonoid abundant in citrus fruits, is recognized for its anti-inflammatory properties and ability to protect against various inflammatory diseases, including drug-induced liver injury. However, the exact effects of naringin on AIH and the mechanisms involved remain poorly understood. PURPOSE: We aim to determine the role of naringin in AIH, exploring its targets and actions in this disease. METHODS: Network pharmacology, molecular docking, and molecular dynamics simulations were utilized to predict the HUB targets connecting naringin, T cell-mediated autoimmune disorders, and AIH. Cellular thermal shift assays were used to determine the binding abilities of naringin with the HUB targets. An in vivo experiment confirmed the impact of naringin treatment on AIH development and underlying mechanisms. RESULTS: Naringin demonstrated therapeutic effects on ConA-induced AIH. There were 455 shared targets between naringin, T cell-mediated autoimmune diseases, and AIH. Ten HUB genes (AKT1, ALB, IL-6, IL-1ß, CTNNB1, TNF, TP53, MAPK3, VEGFA, and JUN) were identified through the PPI network. Gene ontology analysis revealed involvement in gene expression regulation, lipopolysaccharide-mediated signaling, and I-kappa kinase/NFκB signaling. Pathway analysis suggested TNF, Th1/Th2 cell differentiation, and Toll-like receptor pathways, with favorable naringin-HUB gene binding. Molecular docking confirmed albumin (ALB), IL-1ß, IL-6, and TNF as primary targets for naringin. Molecular dynamics simulations showed stable binding in ALB-naringin, TNF-naringin, and IL-1ß-naringin complexes. Naringin's hepatoprotective effect on AIH was supported by increased serum ALB and decreased hepatic inflammatory cytokines including IL-1ß, IL-6, and TNF-α. CONCLUSION: Our data underscore the potential of naringin as a preventive or therapeutical agent in T cell-mediated autoimmune diseases including AIH.


Asunto(s)
Flavanonas , Hepatitis Autoinmune , Simulación del Acoplamiento Molecular , Flavanonas/farmacología , Flavanonas/química , Hepatitis Autoinmune/tratamiento farmacológico , Animales , Citrus/química , Simulación de Dinámica Molecular , Hígado/efectos de los fármacos , Antiinflamatorios/farmacología , Antiinflamatorios/química , Masculino , Farmacología en Red , Concanavalina A , Ratones , Humanos , Linfocitos T/efectos de los fármacos
6.
Water Res X ; 23: 100224, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38711798

RESUMEN

The ongoing evolution of SARS-CoV-2 is a significant concern, especially with the decrease in clinical sequencing efforts, which impedes the ability of public health sectors to prepare for the emergence of new variants and potential COVID-19 outbreaks. Wastewater-based epidemiology (WBE) has been proposed as a surveillance program to detect and monitor the SARS-CoV-2 variants being transmitted in communities. However, research is limited in evaluating the effectiveness of wastewater collection at sentinel sites for monitoring disease prevalence and variant dynamics, especially in terms of inferring the epidemic patterns on a broader scale, such as at the state/province level. This study utilized a multiplexed tiling amplicon-based sequencing (ATOPlex) to track the longitudinal dynamics of variant of concern (VOC) in wastewater collected from municipalities in Queensland, Australia, spanning from 2020 to 2022. We demonstrated that wastewater epidemiology measured by ATOPlex exhibited a strong and consistent correlation with the number of daily confirmed cases. The VOC dynamics observed in wastewater closely aligned with the dynamic profile reported by clinical sequencing. Wastewater sequencing has the potential to provide early warning information for emerging variants. These findings suggest that WBE at sentinel sites, coupled with sensitive sequencing methods, provides a reliable and long-term disease surveillance strategy.

7.
Nat Commun ; 15(1): 4170, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755186

RESUMEN

Endothelial cells are a heterogeneous population with various organ-specific and conserved functions that are critical to organ development, function, and regeneration. Here we report a Sox17-Erg direct reprogramming approach that uses cardiac fibroblasts to create differentiated endothelial cells that demonstrate endothelial-like molecular and physiological functions in vitro and in vivo. Injection of these induced endothelial cells into myocardial infarct sites after injury results in improved vascular perfusion of the scar region. Furthermore, we use genomic analyses to illustrate that Sox17-Erg reprogramming instructs cardiac fibroblasts toward an arterial-like identity. This results in a more efficient direct conversion of fibroblasts into endothelial-like cells when compared to traditional Etv2-based reprogramming. Overall, this Sox17-Erg direct reprogramming strategy offers a robust tool to generate endothelial cells both in vitro and in vivo, and has the potential to be used in repairing injured tissue.


Asunto(s)
Reprogramación Celular , Células Endoteliales , Fibroblastos , Factores de Transcripción SOXF , Regulador Transcripcional ERG , Animales , Ratones , Diferenciación Celular , Reprogramación Celular/genética , Células Endoteliales/metabolismo , Células Endoteliales/citología , Fibroblastos/metabolismo , Fibroblastos/citología , Proteínas HMGB/metabolismo , Proteínas HMGB/genética , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Miocardio/citología , Miocardio/metabolismo , Factores de Transcripción SOXF/metabolismo , Factores de Transcripción SOXF/genética , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
8.
PLoS One ; 19(7): e0305275, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38950026

RESUMEN

Acute compartment syndrome (ACS) is a syndrome in which local circulation is affected due to increased pressure within the compartment. We previously found in patients with calf fractures, the pressure of fascial compartment could be sharply reduced upon the appearance of tension blisters. Deep fascia, as the important structure for compartment, might play key role in this process. Therefore, the aim of the present study was to examine the differences in gene profile in deep fascia tissue in fracture patients of the calf with or without tension blisters, and to explore the role of fascia in pressure improvement in ACS. Patients with lower leg fracture were enrolled and divided into control group (CON group, n = 10) without tension blister, and tension blister group (TB group, n = 10). Deep fascia tissues were collected and LC-MS/MS label-free quantitative proteomics were performed. Genes involved in fascia structure and fibroblast function were further validated by Western blot. The differentially expressed proteins were found to be mainly enriched in pathways related to protein synthesis and processing, stress fiber assembly, cell-substrate adhesion, leukocyte mediated cytotoxicity, and cellular response to stress. Compared with the CON group, the expression of Peroxidasin homolog (PXDN), which promotes the function of fibroblasts, and Leukocyte differentiation antigen 74 (CD74), which enhances the proliferation of fibroblasts, were significantly upregulated (p all <0.05), while the expression of Matrix metalloproteinase-9 (MMP9), which is involved in collagen hydrolysis, and Neutrophil elastase (ELANE), which is involved in elastin hydrolysis, were significantly reduced in the TB group (p all <0.05), indicating fascia tissue underwent microenvironment reconstruction during ACS. In summary, the ACS accompanied by blisters is associated with the enhanced function and proliferation of fibroblasts and reduced hydrolysis of collagen and elastin. The adaptive alterations in the stiffness and elasticity of the deep fascia might be crucial for pressure release of ACS.


Asunto(s)
Síndromes Compartimentales , Fascia , Proteómica , Humanos , Proteómica/métodos , Síndromes Compartimentales/metabolismo , Masculino , Fascia/metabolismo , Fascia/patología , Persona de Mediana Edad , Adulto , Femenino , Enfermedad Aguda , Anciano
9.
Adv Sci (Weinh) ; 11(12): e2304342, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38229183

RESUMEN

Immunotherapy targeting PD-L1 is still ineffective for a wide variety of tumors with high unpredictability. Deploying combined immunotherapy with alternative targeting is practical to overcome this therapeutic resistance. Here, the deficiency of serine-threonine kinase STK24 is observed in tumor cells causing substantial attenuation of tumor growth in murine syngeneic models, a process relying on cytotoxic CD8+ T and NK cells. Mechanistically, STK24 in tumor cells associates with and directly phosphorylates AKT at Thr21, which promotes AKT activation and subsequent PD-L1 induction. Deletion or inhibition of STK24, by contrast, blocks IFN-γ-mediated PD-L1 expression. Various murine models indicate that in vivo silencing of STK24 can significantly enhance the efficacy of the anti-PD-1 blockade strategy. Elevated STK24 levels are observed in patient specimens in multiple tumor types and inversely correlated with intratumoral infiltration of cytotoxic CD8+ T cells and with patient survival. The study collectively identifies STK24 as a critical modulator of antitumor immunity, which engages in AKT and PD-L1/PD-1 signaling and is a promising target for combined immunotherapy.


Asunto(s)
Antígeno B7-H1 , Linfocitos T CD8-positivos , Humanos , Animales , Ratones , Antígeno B7-H1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Escape del Tumor , Línea Celular Tumoral
10.
Nat Cardiovasc Res ; 2(11): 1060-1077, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38524149

RESUMEN

Direct reprogramming of fibroblasts into induced cardiomyocytes holds great promise for heart regeneration. Although considerable progress has been made in understanding the transcriptional and epigenetic mechanisms of iCM reprogramming, its translational regulation remains largely unexplored. Here, we characterized the translational landscape of iCM reprogramming through integrative ribosome and transcriptomic profiling, and found extensive translatome repatterning during this process. Loss of function screening for translational regulators uncovered Ybx1 as a critical barrier to iCM induction. In a mouse model of myocardial infarction, removing Ybx1 enhanced in vivo reprogramming, resulting in improved heart function and reduced scar size. Mechanistically, Ybx1 depletion de-repressed the translation of its direct targets SRF and Baf60c, both of which mediated the effect of Ybx1 depletion on iCM generation. Furthermore, removal of Ybx1 allowed single factor Tbx5-mediated iCM conversion. In summary, this study revealed a new layer of regulatory mechanism that controls cardiac reprogramming at the translational level.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA