Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 986
Filtrar
1.
Annu Rev Cell Dev Biol ; 39: 91-121, 2023 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-37418774

RESUMEN

Intercellular signaling molecules, known as morphogens, act at a long range in developing tissues to provide spatial information and control properties such as cell fate and tissue growth. The production, transport, and removal of morphogens shape their concentration profiles in time and space. Downstream signaling cascades and gene regulatory networks within cells then convert the spatiotemporal morphogen profiles into distinct cellular responses. Current challenges are to understand the diverse molecular and cellular mechanisms underlying morphogen gradient formation, as well as the logic of downstream regulatory circuits involved in morphogen interpretation. This knowledge, combining experimental and theoretical results, is essential to understand emerging properties of morphogen-controlled systems, such as robustness and scaling.

2.
Mol Cell ; 83(9): 1462-1473.e5, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37116493

RESUMEN

DNA binding domains (DBDs) of transcription factors (TFs) recognize DNA sequence motifs that are highly abundant in genomes. Within cells, TFs bind a subset of motif-containing sites as directed by either their DBDs or DBD-external (nonDBD) sequences. To define the relative roles of DBDs and nonDBDs in directing binding preferences, we compared the genome-wide binding of 48 (∼30%) budding yeast TFs with their DBD-only, nonDBD-truncated, and nonDBD-only mutants. With a few exceptions, binding locations differed between DBDs and TFs, resulting from the cumulative action of multiple determinants mapped mostly to disordered nonDBD regions. Furthermore, TFs' preferences for promoters of the fuzzy nucleosome architecture were lost in DBD-only mutants, whose binding spread across promoters, implicating nonDBDs' preferences in this hallmark of budding yeast regulatory design. We conclude that DBDs and nonDBDs employ complementary DNA-targeting strategies, whose balance defines TF binding specificity along genomes.


Asunto(s)
ADN , Factores de Transcripción , Sitios de Unión , Factores de Transcripción/metabolismo , Unión Proteica , ADN/genética
3.
Mol Cell ; 82(2): 248-259, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35063095

RESUMEN

While measurements of RNA expression have dominated the world of single-cell analyses, new single-cell techniques increasingly allow collection of different data modalities, measuring different molecules, structural connections, and intermolecular interactions. Integrating the resulting multimodal single-cell datasets is a new bioinformatics challenge. Equally important, it is a new experimental design challenge for the bench scientist, who is not only choosing from a myriad of techniques for each data modality but also faces new challenges in experimental design. The ultimate goal is to design, execute, and analyze multimodal single-cell experiments that are more than just descriptive but enable the learning of new causal and mechanistic biology. This objective requires strict consideration of the goals behind the analysis, which might range from mapping the heterogeneity of a cellular population to assembling system-wide causal networks that can further our understanding of cellular functions and eventually lead to models of tissues and organs. We review steps and challenges toward this goal. Single-cell transcriptomics is now a mature technology, and methods to measure proteins, lipids, small-molecule metabolites, and other molecular phenotypes at the single-cell level are rapidly developing. Integrating these single-cell readouts so that each cell has measurements of multiple types of data, e.g., transcriptomes, proteomes, and metabolomes, is expected to allow identification of highly specific cellular subpopulations and to provide the basis for inferring causal biological mechanisms.


Asunto(s)
Biología Computacional , Proyectos de Investigación , Análisis de la Célula Individual , Integración de Sistemas , Animales , Perfilación de la Expresión Génica , Humanos , Metabolómica , Proteómica
4.
Annu Rev Neurosci ; 43: 1-30, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31299170

RESUMEN

Cortical interneurons display striking differences in shape, physiology, and other attributes, challenging us to appropriately classify them. We previously suggested that interneuron types should be defined by their role in cortical processing. Here, we revisit the question of how to codify their diversity based upon their division of labor and function as controllers of cortical information flow. We suggest that developmental trajectories provide a guide for appreciating interneuron diversity and argue that subtype identity is generated using a configurational (rather than combinatorial) code of transcription factors that produce attractor states in the underlying gene regulatory network. We present our updated three-stage model for interneuron specification: an initial cardinal step, allocating interneurons into a few major classes, followed by definitive refinement, creating subclasses upon settling within the cortex, and lastly, state determination, reflecting the incorporation of interneurons into functional circuit ensembles. We close by discussing findings indicating that major interneuron classes are both evolutionarily ancient and conserved. We propose that the complexity of cortical circuits is generated by phylogenetically old interneuron types, complemented by an evolutionary increase in principal neuron diversity. This suggests that a natural neurobiological definition of interneuron types might be derived from a match between their developmental origin and computational function.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebral/fisiología , Interneuronas/fisiología , Neurogénesis/fisiología , Animales , Humanos , Neuronas/fisiología , Factores de Transcripción/metabolismo
5.
Annu Rev Cell Dev Biol ; 30: 647-75, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25288119

RESUMEN

Mouse embryonic stem (ES) cells perpetuate in vitro the broad developmental potential of naïve founder cells in the preimplantation embryo. ES cells self-renew relentlessly in culture but can reenter embryonic development seamlessly, differentiating on schedule to form all elements of the fetus. Here we review the properties of these remarkable cells. Arising from the stability, homogeneity, and equipotency of ES cells, we consider the concept of a pluripotent ground state. We evaluate the authenticity of ES cells in relation to cells in the embryo and examine their utility for dissecting mechanisms that confer pluripotency and that execute fate choice. We summarize current knowledge of the transcription factor circuitry that governs the ES cell state and discuss the opportunity to expose molecular logic further through iterative computational modeling and experimentation. Finally, we present a perspective on unresolved questions, including the challenge of deriving ground state pluripotent stem cells from non-rodent species.


Asunto(s)
Células Madre Embrionarias/citología , Animales , División Celular Asimétrica , Blastocisto/citología , Técnicas de Cultivo de Célula , Linaje de la Célula , Células Cultivadas , Reprogramación Celular , Técnicas de Cocultivo , Medios de Cultivo , Medio de Cultivo Libre de Suero , Células Madre de Carcinoma Embrionario/citología , Células Madre Embrionarias/fisiología , Fibroblastos/citología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Estratos Germinativos/citología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Factor Inhibidor de Leucemia/fisiología , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Factores de Transcripción/farmacología , Factores de Transcripción/fisiología
6.
Mol Cell ; 78(5): 960-974.e11, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32330456

RESUMEN

Dynamic cellular processes such as differentiation are driven by changes in the abundances of transcription factors (TFs). However, despite years of studies, our knowledge about the protein copy number of TFs in the nucleus is limited. Here, by determining the absolute abundances of 103 TFs and co-factors during the course of human erythropoiesis, we provide a dynamic and quantitative scale for TFs in the nucleus. Furthermore, we establish the first gene regulatory network of cell fate commitment that integrates temporal protein stoichiometry data with mRNA measurements. The model revealed quantitative imbalances in TFs' cross-antagonistic relationships that underlie lineage determination. Finally, we made the surprising discovery that, in the nucleus, co-repressors are dramatically more abundant than co-activators at the protein level, but not at the RNA level, with profound implications for understanding transcriptional regulation. These analyses provide a unique quantitative framework to understand transcriptional regulation of cell differentiation in a dynamic context.


Asunto(s)
Eritropoyesis/genética , Redes Reguladoras de Genes/genética , Factores de Transcripción/genética , Bases de Datos Factuales , Regulación de la Expresión Génica/genética , Hematopoyesis/genética , Humanos , Proteómica/métodos , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo
7.
Development ; 151(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38958075

RESUMEN

Development is regulated by coordinated changes in gene expression. Control of these changes in expression is largely governed by the binding of transcription factors to specific regulatory elements. However, the packaging of DNA into chromatin prevents the binding of many transcription factors. Pioneer factors overcome this barrier owing to unique properties that enable them to bind closed chromatin, promote accessibility and, in so doing, mediate binding of additional factors that activate gene expression. Because of these properties, pioneer factors act at the top of gene-regulatory networks and drive developmental transitions. Despite the ability to bind target motifs in closed chromatin, pioneer factors have cell type-specific chromatin occupancy and activity. Thus, developmental context clearly shapes pioneer-factor function. Here, we discuss this reciprocal interplay between pioneer factors and development: how pioneer factors control changes in cell fate and how cellular environment influences pioneer-factor binding and activity.


Asunto(s)
Cromatina , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción , Animales , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Cromatina/metabolismo , Humanos , Redes Reguladoras de Genes , Unión Proteica
8.
Proc Natl Acad Sci U S A ; 121(16): e2313440121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38578985

RESUMEN

Developmental phenotypic changes can evolve under selection imposed by age- and size-related ecological differences. Many of these changes occur through programmed alterations to gene expression patterns, but the molecular mechanisms and gene-regulatory networks underlying these adaptive changes remain poorly understood. Many venomous snakes, including the eastern diamondback rattlesnake (Crotalus adamanteus), undergo correlated changes in diet and venom expression as snakes grow larger with age, providing models for identifying mechanisms of timed expression changes that underlie adaptive life history traits. By combining a highly contiguous, chromosome-level genome assembly with measures of expression, chromatin accessibility, and histone modifications, we identified cis-regulatory elements and trans-regulatory factors controlling venom ontogeny in the venom glands of C. adamanteus. Ontogenetic expression changes were significantly correlated with epigenomic changes within genes, immediately adjacent to genes (e.g., promoters), and more distant from genes (e.g., enhancers). We identified 37 candidate transcription factors (TFs), with the vast majority being up-regulated in adults. The ontogenetic change is largely driven by an increase in the expression of TFs associated with growth signaling, transcriptional activation, and circadian rhythm/biological timing systems in adults with corresponding epigenomic changes near the differentially expressed venom genes. However, both expression activation and repression contributed to the composition of both adult and juvenile venoms, demonstrating the complexity and potential evolvability of gene regulation for this trait. Overall, given that age-based trait variation is common across the tree of life, we provide a framework for understanding gene-regulatory-network-driven life-history evolution more broadly.


Asunto(s)
Venenos de Crotálidos , Serpientes Venenosas , Animales , Venenos de Crotálidos/genética , Venenos de Crotálidos/metabolismo , Epigenómica , Crotalus/genética , Crotalus/metabolismo
9.
Proc Natl Acad Sci U S A ; 121(18): e2322751121, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38652750

RESUMEN

Organ-specific gene expression datasets that include hundreds to thousands of experiments allow the reconstruction of organ-level gene regulatory networks (GRNs). However, creating such datasets is greatly hampered by the requirements of extensive and tedious manual curation. Here, we trained a supervised classification model that can accurately classify the organ-of-origin for a plant transcriptome. This K-Nearest Neighbor-based multiclass classifier was used to create organ-specific gene expression datasets for the leaf, root, shoot, flower, and seed in Arabidopsis thaliana. A GRN inference approach was used to determine the: i. influential transcription factors (TFs) in each organ and, ii. most influential TFs for specific biological processes in that organ. These genome-wide, organ-delimited GRNs (OD-GRNs), recalled many known regulators of organ development and processes operating in those organs. Importantly, many previously unknown TF regulators were uncovered as potential regulators of these processes. As a proof-of-concept, we focused on experimentally validating the predicted TF regulators of lipid biosynthesis in seeds, an important food and biofuel trait. Of the top 20 predicted TFs, eight are known regulators of seed oil content, e.g., WRI1, LEC1, FUS3. Importantly, we validated our prediction of MybS2, TGA4, SPL12, AGL18, and DiV2 as regulators of seed lipid biosynthesis. We elucidated the molecular mechanism of MybS2 and show that it induces purple acid phosphatase family genes and lipid synthesis genes to enhance seed lipid content. This general approach has the potential to be extended to any species with sufficiently large gene expression datasets to find unique regulators of any trait-of-interest.


Asunto(s)
Arabidopsis , Regulación de la Expresión Génica de las Plantas , Redes Reguladoras de Genes , Factores de Transcripción , Arabidopsis/genética , Arabidopsis/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Especificidad de Órganos/genética , Transcriptoma/genética , Semillas/genética , Semillas/metabolismo , Perfilación de la Expresión Génica/métodos
10.
Trends Genet ; 39(7): 528-530, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37024335

RESUMEN

Marine larvae have factored heavily in pursuits to understand the origin and evolution of animal life cycles. Recent comparisons of gene expression and chromatin state in different species of sea urchin and annelid show how evolutionary changes in embryonic gene regulation can lead to markedly different larval forms.


Asunto(s)
Estadios del Ciclo de Vida , Erizos de Mar , Animales , Larva/genética , Estadios del Ciclo de Vida/genética , Erizos de Mar/genética
11.
Development ; 150(22)2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37902109

RESUMEN

Multinucleated cells, or syncytia, are found in diverse taxa. Their biological function is often associated with the compartmentalization of biochemical or cellular activities within the syncytium. How such compartments are generated and maintained is poorly understood. The sea urchin embryonic skeleton is secreted by a syncytium, and local patterns of skeletal growth are associated with distinct sub-domains of gene expression within the syncytium. For such molecular compartments to be maintained and to control local patterns of skeletal growth: (1) the mobility of TFs must be restricted to produce stable differences in the transcriptional states of nuclei within the syncytium; and (2) the mobility of biomineralization proteins must also be restricted to produce regional differences in skeletal growth. To test these predictions, we expressed fluorescently tagged forms of transcription factors and biomineralization proteins in sub-domains of the skeletogenic syncytium. We found that both classes of proteins have restricted mobility within the syncytium and identified motifs that limit their mobility. Our findings have general implications for understanding the functional and molecular compartmentalization of syncytia.


Asunto(s)
Erizos de Mar , Factores de Transcripción , Animales , Factores de Transcripción/metabolismo , Células Gigantes/metabolismo , Mesodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica
12.
Brief Bioinform ; 25(5)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39082651

RESUMEN

Constructing accurate gene regulatory network s (GRNs), which reflect the dynamic governing process between genes, is critical to understanding the diverse cellular process and unveiling the complexities in biological systems. With the development of computer sciences, computational-based approaches have been applied to the GRNs inference task. However, current methodologies face challenges in effectively utilizing existing topological information and prior knowledge of gene regulatory relationships, hindering the comprehensive understanding and accurate reconstruction of GRNs. In response, we propose a novel graph neural network (GNN)-based Multi-Task Learning framework for GRN reconstruction, namely MTLGRN. Specifically, we first encode the gene promoter sequences and the gene biological features and concatenate the corresponding feature representations. Then, we construct a multi-task learning framework including GRN reconstruction, Gene knockout predict, and Gene expression matrix reconstruction. With joint training, MTLGRN can optimize the gene latent representations by integrating gene knockout information, promoter characteristics, and other biological attributes. Extensive experimental results demonstrate superior performance compared with state-of-the-art baselines on the GRN reconstruction task, efficiently leveraging biological knowledge and comprehensively understanding the gene regulatory relationships. MTLGRN also pioneered attempts to simulate gene knockouts on bulk data by incorporating gene knockout information.


Asunto(s)
Biología Computacional , Redes Reguladoras de Genes , Biología Computacional/métodos , Técnicas de Inactivación de Genes , Redes Neurales de la Computación , Humanos , Regiones Promotoras Genéticas , Algoritmos
13.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38886006

RESUMEN

Reconstructing the topology of gene regulatory network from gene expression data has been extensively studied. With the abundance functional transcriptomic data available, it is now feasible to systematically decipher regulatory interaction dynamics in a logic form such as a Boolean network (BN) framework, which qualitatively indicates how multiple regulators aggregated to affect a common target gene. However, inferring both the network topology and gene interaction dynamics simultaneously is still a challenging problem since gene expression data are typically noisy and data discretization is prone to information loss. We propose a new method for BN inference from time-series transcriptional profiles, called LogicGep. LogicGep formulates the identification of Boolean functions as a symbolic regression problem that learns the Boolean function expression and solve it efficiently through multi-objective optimization using an improved gene expression programming algorithm. To avoid overly emphasizing dynamic characteristics at the expense of topology structure ones, as traditional methods often do, a set of promising Boolean formulas for each target gene is evolved firstly, and a feed-forward neural network trained with continuous expression data is subsequently employed to pick out the final solution. We validated the efficacy of LogicGep using multiple datasets including both synthetic and real-world experimental data. The results elucidate that LogicGep adeptly infers accurate BN models, outperforming other representative BN inference algorithms in both network topology reconstruction and the identification of Boolean functions. Moreover, the execution of LogicGep is hundreds of times faster than other methods, especially in the case of large network inference.


Asunto(s)
Algoritmos , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Perfilación de la Expresión Génica/métodos , Humanos , Transcriptoma , Programas Informáticos , Biología Computacional/métodos , Redes Neurales de la Computación
14.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38581416

RESUMEN

The inference of gene regulatory networks (GRNs) from gene expression profiles has been a key issue in systems biology, prompting many researchers to develop diverse computational methods. However, most of these methods do not reconstruct directed GRNs with regulatory types because of the lack of benchmark datasets or defects in the computational methods. Here, we collect benchmark datasets and propose a deep learning-based model, DeepFGRN, for reconstructing fine gene regulatory networks (FGRNs) with both regulation types and directions. In addition, the GRNs of real species are always large graphs with direction and high sparsity, which impede the advancement of GRN inference. Therefore, DeepFGRN builds a node bidirectional representation module to capture the directed graph embedding representation of the GRN. Specifically, the source and target generators are designed to learn the low-dimensional dense embedding of the source and target neighbors of a gene, respectively. An adversarial learning strategy is applied to iteratively learn the real neighbors of each gene. In addition, because the expression profiles of genes with regulatory associations are correlative, a correlation analysis module is designed. Specifically, this module not only fully extracts gene expression features, but also captures the correlation between regulators and target genes. Experimental results show that DeepFGRN has a competitive capability for both GRN and FGRN inference. Potential biomarkers and therapeutic drugs for breast cancer, liver cancer, lung cancer and coronavirus disease 2019 are identified based on the candidate FGRNs, providing a possible opportunity to advance our knowledge of disease treatments.


Asunto(s)
Redes Reguladoras de Genes , Neoplasias Hepáticas , Humanos , Biología de Sistemas/métodos , Transcriptoma , Algoritmos , Biología Computacional/métodos
15.
Brief Bioinform ; 25(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38261340

RESUMEN

The recent advances of single-cell RNA sequencing (scRNA-seq) have enabled reliable profiling of gene expression at the single-cell level, providing opportunities for accurate inference of gene regulatory networks (GRNs) on scRNA-seq data. Most methods for inferring GRNs suffer from the inability to eliminate transitive interactions or necessitate expensive computational resources. To address these, we present a novel method, termed GMFGRN, for accurate graph neural network (GNN)-based GRN inference from scRNA-seq data. GMFGRN employs GNN for matrix factorization and learns representative embeddings for genes. For transcription factor-gene pairs, it utilizes the learned embeddings to determine whether they interact with each other. The extensive suite of benchmarking experiments encompassing eight static scRNA-seq datasets alongside several state-of-the-art methods demonstrated mean improvements of 1.9 and 2.5% over the runner-up in area under the receiver operating characteristic curve (AUROC) and area under the precision-recall curve (AUPRC). In addition, across four time-series datasets, maximum enhancements of 2.4 and 1.3% in AUROC and AUPRC were observed in comparison to the runner-up. Moreover, GMFGRN requires significantly less training time and memory consumption, with time and memory consumed <10% compared to the second-best method. These findings underscore the substantial potential of GMFGRN in the inference of GRNs. It is publicly available at https://github.com/Lishuoyy/GMFGRN.


Asunto(s)
Benchmarking , Redes Reguladoras de Genes , Área Bajo la Curva , Aprendizaje , Redes Neurales de la Computación
16.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38935070

RESUMEN

Inferring gene regulatory network (GRN) is one of the important challenges in systems biology, and many outstanding computational methods have been proposed; however there remains some challenges especially in real datasets. In this study, we propose Directed Graph Convolutional neural network-based method for GRN inference (DGCGRN). To better understand and process the directed graph structure data of GRN, a directed graph convolutional neural network is conducted which retains the structural information of the directed graph while also making full use of neighbor node features. The local augmentation strategy is adopted in graph neural network to solve the problem of poor prediction accuracy caused by a large number of low-degree nodes in GRN. In addition, for real data such as E.coli, sequence features are obtained by extracting hidden features using Bi-GRU and calculating the statistical physicochemical characteristics of gene sequence. At the training stage, a dynamic update strategy is used to convert the obtained edge prediction scores into edge weights to guide the subsequent training process of the model. The results on synthetic benchmark datasets and real datasets show that the prediction performance of DGCGRN is significantly better than existing models. Furthermore, the case studies on bladder uroepithelial carcinoma and lung cancer cells also illustrate the performance of the proposed model.


Asunto(s)
Biología Computacional , Redes Reguladoras de Genes , Redes Neurales de la Computación , Humanos , Biología Computacional/métodos , Algoritmos , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Escherichia coli/genética
17.
Trends Immunol ; 44(7): 530-541, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37258360

RESUMEN

Specific combinations of transcription factors (TFs) control the gene expression programs that underlie specialized immune responses. Previous models of TF function in immunocytes had restricted each TF to a single functional categorization [e.g., lineage-defining (LDTFs) vs. signal-dependent TFs (SDTFs)] within one cell type. Synthesizing recent results, we instead propose a variegated model of immunological TF function, whereby many TFs have flexible and different roles across distinct cell states, contributing to cell phenotypic diversity. We discuss evidence in support of this variegated model, describe contextual inputs that enable TF diversification, and look to the future to imagine warranted experimental and computational tools to build quantitative and predictive models of immunocyte gene regulatory networks.


Asunto(s)
Regulación de la Expresión Génica , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Redes Reguladoras de Genes , Sistema Inmunológico/metabolismo
18.
Proc Natl Acad Sci U S A ; 120(2): e2212151120, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36608289

RESUMEN

Cells cope with and adapt to ever-changing environmental conditions. Sophisticated regulatory networks allow cells to adjust to these fluctuating environments. One such archetypal system is the Saccharomyces cerevisiae Pho regulon. When external inorganic phosphate (Pi) concentration is low, the Pho regulon activates, expressing genes that scavenge external and internal Pi. However, the precise mechanism controlling this regulon remains elusive. We conducted a systems analysis of the Pho regulon on the single-cell level under well-controlled environmental conditions. This analysis identified a robust, perfectly adapted Pho regulon state in intermediate Pi conditions, and we identified an intermediate nuclear localization state of the transcriptional master regulator Pho4p. The existence of an intermediate nuclear Pho4p state unifies and resolves outstanding incongruities associated with the Pho regulon, explains the observed programmatic states of the Pho regulon, and improves our general understanding of how nature evolves and controls sophisticated gene regulatory networks. We further propose that robustness and perfect adaptation are not achieved through complex network-centric control but by simple transport biophysics. The ubiquity of multitransporter systems suggests that similar mechanisms could govern the function of other regulatory networks as well.


Asunto(s)
Fosfatos , Saccharomyces cerevisiae , Fosfatos/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Regulón/genética , Aclimatación , Regulación Bacteriana de la Expresión Génica , Proteínas Bacterianas/metabolismo
19.
Proc Natl Acad Sci U S A ; 120(5): e2214883120, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36706221

RESUMEN

Sex peptide (SP), a seminal fluid protein of Drosophila melanogaster males, has been described as driving a virgin-to-mated switch in females, through eliciting an array of responses including increased egg laying, activity, and food intake and a decreased remating rate. While it is known that SP achieves this, at least in part, by altering neuronal signaling in females, the genetic architecture and temporal dynamics of the female's response to SP remain elusive. We used a high-resolution time series RNA-sequencing dataset of female heads at 10 time points within the first 24 h after mating to learn about the genetic architecture, at the gene and exon levels, of the female's response to SP. We find that SP is not essential to trigger early aspects of a virgin-to-mated transcriptional switch, which includes changes in a metabolic gene regulatory network. However, SP is needed to maintain and diversify metabolic changes and to trigger changes in a neuronal gene regulatory network. We further find that SP alters rhythmic gene expression in females and suggests that SP's disruption of the female's circadian rhythm might be key to its widespread effects.


Asunto(s)
Relojes Circadianos , Proteínas de Drosophila , Animales , Masculino , Femenino , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Espermatozoides/metabolismo , Relojes Circadianos/genética , Factores de Tiempo , Péptidos/metabolismo , Perfilación de la Expresión Génica , Conducta Sexual Animal/fisiología
20.
Plant J ; 118(5): 1668-1688, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38407828

RESUMEN

Bioenergy sorghum is a low-input, drought-resilient, deep-rooting annual crop that has high biomass yield potential enabling the sustainable production of biofuels, biopower, and bioproducts. Bioenergy sorghum's 4-5 m stems account for ~80% of the harvested biomass. Stems accumulate high levels of sucrose that could be used to synthesize bioethanol and useful biopolymers if information about cell-type gene expression and regulation in stems was available to enable engineering. To obtain this information, laser capture microdissection was used to isolate and collect transcriptome profiles from five major cell types that are present in stems of the sweet sorghum Wray. Transcriptome analysis identified genes with cell-type-specific and cell-preferred expression patterns that reflect the distinct metabolic, transport, and regulatory functions of each cell type. Analysis of cell-type-specific gene regulatory networks (GRNs) revealed that unique transcription factor families contribute to distinct regulatory landscapes, where regulation is organized through various modes and identifiable network motifs. Cell-specific transcriptome data was combined with known secondary cell wall (SCW) networks to identify the GRNs that differentially activate SCW formation in vascular sclerenchyma and epidermal cells. The spatial transcriptomic dataset provides a valuable source of information about the function of different sorghum cell types and GRNs that will enable the engineering of bioenergy sorghum stems, and an interactive web application developed during this project will allow easy access and exploration of the data (https://mc-lab.shinyapps.io/lcm-dataset/).


Asunto(s)
Biocombustibles , Pared Celular , Regulación de la Expresión Génica de las Plantas , Redes Reguladoras de Genes , Tallos de la Planta , Sorghum , Transcriptoma , Sorghum/genética , Sorghum/metabolismo , Tallos de la Planta/genética , Tallos de la Planta/metabolismo , Pared Celular/metabolismo , Pared Celular/genética , Perfilación de la Expresión Génica
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda