Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Mol Cell ; 82(7): 1297-1312.e8, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35219381

RESUMEN

Synthetic lethality through combinatorial targeting DNA damage response (DDR) pathways provides exciting anticancer therapeutic benefit. Currently, the long noncoding RNAs (lncRNAs) have been implicated in tumor drug resistance; however, their potential significance in DDR is still largely unknown. Here, we report that a human lncRNA, CTD-2256P15.2, encodes a micropeptide, named PAR-amplifying and CtIP-maintaining micropeptide (PACMP), with a dual function to maintain CtIP abundance and promote poly(ADP-ribosyl)ation. PACMP not only prevents CtIP from ubiquitination through inhibiting the CtIP-KLHL15 association but also directly binds DNA damage-induced poly(ADP-ribose) chains to enhance PARP1-dependent poly(ADP-ribosyl)ation. Targeting PACMP alone inhibits tumor growth by causing a synthetic lethal interaction between CtIP and PARP inhibitions and confers sensitivity to PARP/ATR/CDK4/6 inhibitors, ionizing radiation, epirubicin, and camptothecin. Our findings reveal that a lncRNA-derived micropeptide regulates cancer progression and drug resistance by modulating DDR, whose inhibition could be employed to augment the existing anticancer therapeutic strategies.


Asunto(s)
Endodesoxirribonucleasas , Neoplasias , Péptidos , Poli ADP Ribosilación , ARN Largo no Codificante , Reparación del ADN , Endodesoxirribonucleasas/metabolismo , Humanos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Péptidos/farmacología , Poli Adenosina Difosfato Ribosa/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
2.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38819253

RESUMEN

Spatially resolved transcriptomics (SRT) has emerged as a powerful tool for investigating gene expression in spatial contexts, providing insights into the molecular mechanisms underlying organ development and disease pathology. However, the expression sparsity poses a computational challenge to integrate other modalities (e.g. histological images and spatial locations) that are simultaneously captured in SRT datasets for spatial clustering and variation analyses. In this study, to meet such a challenge, we propose multi-modal domain adaption for spatial transcriptomics (stMDA), a novel multi-modal unsupervised domain adaptation method, which integrates gene expression and other modalities to reveal the spatial functional landscape. Specifically, stMDA first learns the modality-specific representations from spatial multi-modal data using multiple neural network architectures and then aligns the spatial distributions across modal representations to integrate these multi-modal representations, thus facilitating the integration of global and spatially local information and improving the consistency of clustering assignments. Our results demonstrate that stMDA outperforms existing methods in identifying spatial domains across diverse platforms and species. Furthermore, stMDA excels in identifying spatially variable genes with high prognostic potential in cancer tissues. In conclusion, stMDA as a new tool of multi-modal data integration provides a powerful and flexible framework for analyzing SRT datasets, thereby advancing our understanding of intricate biological systems.


Asunto(s)
Perfilación de la Expresión Génica , Transcriptoma , Humanos , Perfilación de la Expresión Génica/métodos , Análisis por Conglomerados , Biología Computacional/métodos , Redes Neurales de la Computación , Neoplasias/genética , Algoritmos
3.
Brief Bioinform ; 25(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38324623

RESUMEN

Recent advances in spatially resolved transcriptomics (SRT) have brought ever-increasing opportunities to characterize expression landscape in the context of tissue spatiality. Nevertheless, there still exist multiple challenges to accurately detect spatial functional regions in tissue. Here, we present a novel contrastive learning framework, SPAtially Contrastive variational AutoEncoder (SpaCAE), which contrasts transcriptomic signals of each spot and its spatial neighbors to achieve fine-grained tissue structures detection. By employing a graph embedding variational autoencoder and incorporating a deep contrastive strategy, SpaCAE achieves a balance between spatial local information and global information of expression, enabling effective learning of representations with spatial constraints. Particularly, SpaCAE provides a graph deconvolutional decoder to address the smoothing effect of local spatial structure on expression's self-supervised learning, an aspect often overlooked by current graph neural networks. We demonstrated that SpaCAE could achieve effective performance on SRT data generated from multiple technologies for spatial domains identification and data denoising, making it a remarkable tool to obtain novel insights from SRT studies.


Asunto(s)
Perfilación de la Expresión Génica , Transcriptoma , Redes Neurales de la Computación
4.
Brief Bioinform ; 24(5)2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37507114

RESUMEN

Advances in single-cell multi-omics technology provide an unprecedented opportunity to fully understand cellular heterogeneity. However, integrating omics data from multiple modalities is challenging due to the individual characteristics of each measurement. Here, to solve such a problem, we propose a contrastive and generative deep self-expression model, called single-cell multimodal self-expressive integration (scMSI), which integrates the heterogeneous multimodal data into a unified manifold space. Specifically, scMSI first learns each omics-specific latent representation and self-expression relationship to consider the characteristics of different omics data by deep self-expressive generative model. Then, scMSI combines these omics-specific self-expression relations through contrastive learning. In such a way, scMSI provides a paradigm to integrate multiple omics data even with weak relation, which effectively achieves the representation learning and data integration into a unified framework. We demonstrate that scMSI provides a cohesive solution for a variety of analysis tasks, such as integration analysis, data denoising, batch correction and spatial domain detection. We have applied scMSI on various single-cell and spatial multimodal datasets to validate its high effectiveness and robustness in diverse data types and application scenarios.


Asunto(s)
Aprendizaje , Multiómica
5.
Brief Bioinform ; 24(4)2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37253698

RESUMEN

Spatially resolved transcriptomics (SRT) enable the comprehensive characterization of transcriptomic profiles in the context of tissue microenvironments. Unveiling spatial transcriptional heterogeneity needs to effectively incorporate spatial information accounting for the substantial spatial correlation of expression measurements. Here, we develop a computational method, SpaSRL (spatially aware self-representation learning), which flexibly enhances and decodes spatial transcriptional signals to simultaneously achieve spatial domain detection and spatial functional genes identification. This novel tunable spatially aware strategy of SpaSRL not only balances spatial and transcriptional coherence for the two tasks, but also can transfer spatial correlation constraint between them based on a unified model. In addition, this joint analysis by SpaSRL deciphers accurate and fine-grained tissue structures and ensures the effective extraction of biologically informative genes underlying spatial architecture. We verified the superiority of SpaSRL on spatial domain detection, spatial functional genes identification and data denoising using multiple SRT datasets obtained by different platforms and tissue sections. Our results illustrate SpaSRL's utility in flexible integration of spatial information and novel discovery of biological insights from spatial transcriptomic datasets.


Asunto(s)
Perfilación de la Expresión Génica , Aprendizaje , Transcriptoma
6.
Brief Bioinform ; 23(2)2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35079777

RESUMEN

The integration of multi-omics data makes it possible to understand complex biological organisms at the system level. Numerous integration approaches have been developed by assuming a common underlying data space. Due to the noise and heterogeneity of biological data, the performance of these approaches is greatly affected. In this work, we propose a novel deep neural network architecture, named Deep Latent Space Fusion (DLSF), which integrates the multi-omics data by learning consistent manifold in the sample latent space for disease subtypes identification. DLSF is built upon a cycle autoencoder with a shared self-expressive layer, which can naturally and adaptively merge nonlinear features at each omics level into one unified sample manifold and produce adaptive representation of heterogeneous samples at the multi-omics level. We have assessed DLSF on various biological and biomedical datasets to validate its effectiveness. DLSF can efficiently and accurately capture the intrinsic manifold of the sample structures or sample clusters compared with other state-of-the-art methods, and DLSF yielded more significant outcomes for biological significance, survival prognosis and clinical relevance in application of cancer study in The Cancer Genome Atlas. Notably, as a deep case study, we determined a new molecular subtype of kidney renal clear cell carcinoma that may benefit immunotherapy in the viewpoint of multi-omics, and we further found potential subtype-specific biomarkers from multiple omics data, which were validated by independent datasets. In addition, we applied DLSF to identify potential therapeutic agents of different molecular subtypes of chronic lymphocytic leukemia, demonstrating the scalability of DLSF in diverse omics data types and application scenarios.


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética
7.
Opt Express ; 27(13): 18351-18362, 2019 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-31252780

RESUMEN

Anti-reflection (AR) coating is a critical technology and an ongoing challenge for terahertz systems. The subwavelength structure (SWS) is an effective AR method, whereas the current manufacturing techniques, such as chemical etching and ultrafast laser processing, are low-efficient and low-quality for processing structures at the hundred-micron scale on hard brittle materials. We present a study of broadband SWSs directly ablated on the surface of quartz crystal by precisely controlled CO2 laser pulses, instead of commonly used ultra-fast lasers. The processing time of SWS can be shortened by two orders of magnitude compared with that by ultra-fast laser pulses. The SWS samples exhibit excellent AR properties with maximum transmittance of 97% at 0.71 THz, peak transmittance improvement of 13.5%, and optimal efficiency spectrum of 0.28-1.21 THz with transmittance >90%. The AR properties of SWS samples are in agreement with the simulated expectation and exist over a wide range of incidence angles up to ∼40°. The imaging of an object using SWS as the substrate shows an obvious improvement in imaging quality. We present an efficient and practical way to improve the transmission of optical components of materials, such as quartz crystal, alumina, and sapphire, in the terahertz band.

8.
Opt Express ; 27(8): 10705-10728, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-31052925

RESUMEN

In a large-scale, high-power laser facility, fused silica optics plays an irreplaceable role to transmit extremely intense lasers. However, the surface fractures, such as surface pit, crack, and scratch and laser damage site, of fused silica optics will shorten the lifetime of the optics and thus limit the output performance of the laser facility. In this work, besides experimental study, finite difference time domain (FDTD) simulation is performed to study hydrofluoric acid-based (HF-based) etching effect on the surface fractures. The effect of local surface curvature on etching rate is discussed and an explicit local-curvature-dependent etching model is proposed. Based on this model, the result from FDTD simulation qualitatively agrees very well with that of the experiment. It is demonstrated that the FDTD simulation is efficient to predict the morphological evolution of the surface fractures during etching. In addition, it is found that the surface fractures will be passivated and HF-based etching can greatly suppress the laser-damage growth of laser-induced damage to the surface site of fused silica optics.

9.
Bioinformatics ; 33(17): 2706-2714, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28520848

RESUMEN

MOTIVATION: Integrating different omics profiles is a challenging task, which provides a comprehensive way to understand complex diseases in a multi-view manner. One key for such an integration is to extract intrinsic patterns in concordance with data structures, so as to discover consistent information across various data types even with noise pollution. Thus, we proposed a novel framework called 'pattern fusion analysis' (PFA), which performs automated information alignment and bias correction, to fuse local sample-patterns (e.g. from each data type) into a global sample-pattern corresponding to phenotypes (e.g. across most data types). In particular, PFA can identify significant sample-patterns from different omics profiles by optimally adjusting the effects of each data type to the patterns, thereby alleviating the problems to process different platforms and different reliability levels of heterogeneous data. RESULTS: To validate the effectiveness of our method, we first tested PFA on various synthetic datasets, and found that PFA can not only capture the intrinsic sample clustering structures from the multi-omics data in contrast to the state-of-the-art methods, such as iClusterPlus, SNF and moCluster, but also provide an automatic weight-scheme to measure the corresponding contributions by data types or even samples. In addition, the computational results show that PFA can reveal shared and complementary sample-patterns across data types with distinct signal-to-noise ratios in Cancer Cell Line Encyclopedia (CCLE) datasets, and outperforms over other works at identifying clinically distinct cancer subtypes in The Cancer Genome Atlas (TCGA) datasets. AVAILABILITY AND IMPLEMENTATION: PFA has been implemented as a Matlab package, which is available at http://www.sysbio.ac.cn/cb/chenlab/images/PFApackage_0.1.rar . CONTACT: lnchen@sibs.ac.cn , liujuan@whu.edu.cn or zengtao@sibs.ac.cn. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Neoplasias/genética , Análisis por Conglomerados , Humanos , Reproducibilidad de los Resultados , Relación Señal-Ruido , Programas Informáticos
10.
Methods ; 124: 25-35, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28710010

RESUMEN

Transcription factors (TFs) could regulate physiological transitions or determine stable phenotypic diversity. The accurate estimation on TF regulatory signals or functional activities is of great significance to guide biological experiments or elucidate molecular mechanisms, but still remains challenging. Traditional methods identify TF regulatory signals at the population level, which masks heterogeneous regulation mechanisms in individuals or subgroups, thus resulting in inaccurate analyses. Here, we propose a novel computational framework, namely local network component analysis (LNCA), to exploit data heterogeneity and automatically quantify accurate transcription factor activity (TFA) in practical terms, through integrating the partitioned expression sets (i.e., local information) and prior TF-gene regulatory knowledge. Specifically, LNCA adopts an adaptive optimization strategy, which evaluates the local similarities of regulation controls and corrects biases during data integration, to construct the TFA landscape. In particular, we first numerically demonstrate the effectiveness of LNCA for the simulated data sets, compared with traditional methods, such as FastNCA, ROBNCA and NINCA. Then, we apply our model to two real data sets with implicit temporal or spatial regulation variations. The results show that LNCA not only recognizes the periodic mode along the S. cerevisiae cell cycle process, but also substantially outperforms over other methods in terms of accuracy and consistency. In addition, the cross-validation study for glioblastomas multiforme (GBM) indicates that the TFAs, identified by LNCA, can better distinguish clinically distinct tumor groups than the expression values of the corresponding TFs, thus opening a new way to classify tumor subtypes and also providing a novel insight into cancer heterogeneity. AVAILABILITY: LNCA was implemented as a Matlab package, which is available at http://sysbio.sibcb.ac.cn/cb/chenlab/software.htm/LNCApackage_0.1.rar.


Asunto(s)
Algoritmos , Neoplasias Encefálicas/genética , Glioblastoma/genética , Proteínas de Neoplasias/genética , Factores de Transcripción/genética , Transcripción Genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Ciclo Celular/genética , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Glioblastoma/diagnóstico , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Humanos , Pronóstico , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Análisis de Supervivencia , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA