Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 171(5): 1125-1137.e11, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29107333

RESUMEN

Human cytotoxic lymphocytes kill intracellular microbes. The cytotoxic granule granzyme proteases released by cytotoxic lymphocytes trigger oxidative bacterial death by disrupting electron transport, generating superoxide anion and inactivating bacterial oxidative defenses. However, they also cause non-oxidative cell death because anaerobic bacteria are also killed. Here, we use differential proteomics to identify granzyme B substrates in three unrelated bacteria: Escherichia coli, Listeria monocytogenes, and Mycobacteria tuberculosis. Granzyme B cleaves a highly conserved set of proteins in all three bacteria, which function in vital biosynthetic and metabolic pathways that are critical for bacterial survival under diverse environmental conditions. Key proteins required for protein synthesis, folding, and degradation are also substrates, including multiple aminoacyl tRNA synthetases, ribosomal proteins, protein chaperones, and the Clp system. Because killer cells use a multipronged strategy to target vital pathways, bacteria may not easily become resistant to killer cell attack.


Asunto(s)
Escherichia coli/citología , Granzimas/metabolismo , Células Asesinas Naturales/enzimología , Listeria monocytogenes/citología , Mycobacterium tuberculosis/citología , Linfocitos T Citotóxicos/enzimología , Aminoacil-ARNt Sintetasas/metabolismo , Animales , Escherichia coli/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Listeria monocytogenes/metabolismo , Redes y Vías Metabólicas , Ratones , Mycobacterium tuberculosis/metabolismo , Biosíntesis de Proteínas , Proteómica , Ribosomas/metabolismo , Linfocitos T Citotóxicos/inmunología
2.
Bioessays ; 42(9): e2000083, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32638413

RESUMEN

Several metabolites serve as substrates for histone modifications and communicate changes in the metabolic environment to the epigenome. Technologies such as metabolomics and proteomics have allowed us to reconstruct the interactions between metabolic pathways and histones. These technologies have shed light on how nutrient availability can have a dramatic effect on various histone modifications. This metabolism-epigenome cross talk plays a fundamental role in development, immune function, and diseases like cancer. Yet, major challenges remain in understanding the interactions between cellular metabolism and the epigenome. How the levels and fluxes of various metabolites impact epigenetic marks is still unclear. Discussed herein are recent applications and the potential of systems biology methods such as flux tracing and metabolic modeling to address these challenges and to uncover new metabolic-epigenetic interactions. These systems approaches can ultimately help elucidate how nutrients shape the epigenome of microbes and mammalian cells.


Asunto(s)
Código de Histonas , Lectura , Animales , Epigénesis Genética , Epigenómica , Histonas/genética , Histonas/metabolismo , Humanos , Nutrientes
3.
Genome Res ; 27(6): 959-972, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28356321

RESUMEN

Agonistic encounters are powerful effectors of future behavior, and the ability to learn from this type of social challenge is an essential adaptive trait. We recently identified a conserved transcriptional program defining the response to social challenge across animal species, highly enriched in transcription factor (TF), energy metabolism, and developmental signaling genes. To understand the trajectory of this program and to uncover the most important regulatory influences controlling this response, we integrated gene expression data with the chromatin landscape in the hypothalamus, frontal cortex, and amygdala of socially challenged mice over time. The expression data revealed a complex spatiotemporal patterning of events starting with neural signaling molecules in the frontal cortex and ending in the modulation of developmental factors in the amygdala and hypothalamus, underpinned by a systems-wide shift in expression of energy metabolism-related genes. The transcriptional signals were correlated with significant shifts in chromatin accessibility and a network of challenge-associated TFs. Among these, the conserved metabolic and developmental regulator ESRRA was highlighted for an especially early and important regulatory role. Cell-type deconvolution analysis attributed the differential metabolic and developmental signals in this social context primarily to oligodendrocytes and neurons, respectively, and we show that ESRRA is expressed in both cell types. Localizing ESRRA binding sites in cortical chromatin, we show that this nuclear receptor binds both differentially expressed energy-related and neurodevelopmental TF genes. These data link metabolic and neurodevelopmental signaling to social challenge, and identify key regulatory drivers of this process with unprecedented tissue and temporal resolution.


Asunto(s)
Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo , Receptores de Estrógenos/genética , Estrés Psicológico/genética , Factores de Transcripción/genética , Conducta Agonística , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiopatología , Animales , Cromatina/ultraestructura , Metabolismo Energético/genética , Lóbulo Frontal/metabolismo , Lóbulo Frontal/fisiopatología , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Masculino , Ratones , Neuronas/citología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Unión Proteica , Receptores de Estrógenos/metabolismo , Transducción de Señal , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Factores de Transcripción/metabolismo , Transcripción Genética , Receptor Relacionado con Estrógeno ERRalfa
4.
J Nutr ; 150(12): 3075-3085, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-32937657

RESUMEN

BACKGROUND: Alpha-tocopherol (αT), the bioactive constituent of vitamin E, is essential for fertility and neurological development. Synthetic αT (8 stereoisomers; all rac-αT) is added to infant formula at higher concentrations than natural αT (RRR-αT only) to adjust for bio-potency differences, but its effects on brain development are poorly understood. OBJECTIVES: The objective was to determine the impact of bio-potency-adjusted dietary all rac-αT versus RRR-αT, fed to dams, on the hippocampal gene expression in weanling mice. METHODS: Male/female pairs of C57BL/6J mice were fed AIN 93-G containing RRR-αT (NAT) or all rac-αT (SYN) at 37.5 or 75 IU/kg (n = 10/group) throughout gestation and lactation. Male pups were euthanized at 21 days. Half the brain was evaluated for the αT concentration and stereoisomer distribution. The hippocampus was dissected from the other half, and RNA was extracted and sequenced. Milk αT was analyzed in separate dams. RESULTS: A total of 797 differentially expressed genes (DEGs) were identified in the hippocampi across the 4 dietary groups, at a false discovery rate of 10%. Comparing the NAT-37.5 group to the NAT-75 group or the SYN-37.5 group to the SYN-75 group, small differences in brain αT concentrations (10%; P < 0.05) led to subtle changes (<10%) in gene expression of 600 (NAT) or 487 genes (SYN), which were statistically significant. Marked differences in brain αT stereoisomer profiles (P < 0.0001) had a small effect on fewer genes (NAT-37.5 vs. SYN-37.5, 179; NAT-75 vs. SYN-75, 182). Most of the DEGs were involved in transcription regulation and synapse formation. A network analysis constructed around known vitamin E interacting proteins (VIPs) revealed a group of 32 DEGs between NAT-37.5 vs. SYN-37.5, explained by expression of the gene for the VIP, protein kinase C zeta (Pkcz). CONCLUSIONS: In weanling mouse hippocampi, a network of genes involved in transcription regulation and synapse formation was differentially affected by dam diet αT concentration and source: all rac-αT or RRR-αT.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , alfa-Tocoferol/metabolismo , Animales , Dieta , Femenino , Regulación de la Expresión Génica/fisiología , Masculino , Ratones , Leche/química , Leche/metabolismo , alfa-Tocoferol/química
5.
PLoS Genet ; 13(7): e1006840, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28704398

RESUMEN

Animals exhibit dramatic immediate behavioral plasticity in response to social interactions, and brief social interactions can shape the future social landscape. However, the molecular mechanisms contributing to behavioral plasticity are unclear. Here, we show that the genome dynamically responds to social interactions with multiple waves of transcription associated with distinct molecular functions in the brain of male threespined sticklebacks, a species famous for its behavioral repertoire and evolution. Some biological functions (e.g., hormone activity) peaked soon after a brief territorial challenge and then declined, while others (e.g., immune response) peaked hours afterwards. We identify transcription factors that are predicted to coordinate waves of transcription associated with different components of behavioral plasticity. Next, using H3K27Ac as a marker of chromatin accessibility, we show that a brief territorial intrusion was sufficient to cause rapid and dramatic changes in the epigenome. Finally, we integrate the time course brain gene expression data with a transcriptional regulatory network, and link gene expression to changes in chromatin accessibility. This study reveals rapid and dramatic epigenomic plasticity in response to a brief, highly consequential social interaction.


Asunto(s)
Conducta Animal/fisiología , Plasticidad Neuronal/genética , Smegmamorpha/genética , Conducta Social , Transcripción Genética , Animales , Evolución Biológica , Cerebro/fisiología , Cromatina/genética , Diencéfalo/fisiología , Epigenómica , Genoma , Análisis de Secuencia de ARN , Smegmamorpha/fisiología , Factores de Transcripción/genética
6.
BMC Bioinformatics ; 20(1): 307, 2019 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-31182013

RESUMEN

BACKGROUND: The maturation of the female germ cell, the oocyte, requires the synthesis and storing of all the necessary metabolites to support multiple divisions after fertilization. Oocyte maturation is only possible in the presence of surrounding, diverse, and changing layers of somatic cells. Our understanding of metabolic interactions between the oocyte and somatic cells has been limited due to dynamic nature of ovarian follicle development, thus warranting a systems approach. RESULTS: Here, we developed a genome-scale metabolic model of the mouse ovarian follicle. This model was constructed using an updated mouse general metabolic model (Mouse Recon 2) and contains several key ovarian follicle development metabolic pathways. We used this model to characterize the changes in the metabolism of each follicular cell type (i.e., oocyte, granulosa cells, including cumulus and mural cells), during ovarian follicle development in vivo. Using this model, we predicted major metabolic pathways that are differentially active across multiple follicle stages. We identified a set of possible secreted and consumed metabolites that could potentially serve as biomarkers for monitoring follicle development, as well as metabolites for addition to in vitro culture media that support the growth and maturation of primordial follicles. CONCLUSIONS: Our systems approach to model follicle metabolism can guide future experimental studies to validate the model results and improve oocyte maturation approaches and support growth of primordial follicles in vitro.


Asunto(s)
Comunicación Celular , Genoma , Modelos Biológicos , Folículo Ovárico/metabolismo , Animales , Diferenciación Celular , Femenino , Redes y Vías Metabólicas , Ratones , Folículo Ovárico/citología
7.
PLoS Comput Biol ; 14(12): e1006677, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30596642

RESUMEN

Antibiotics need to be effective in diverse environments in vivo. However, the pathogen microenvironment can have a significant impact on antibiotic potency. Further, antibiotics are increasingly used in combinations to combat resistance, yet, the effect of microenvironments on drug-combination efficacy is unknown. To exhaustively explore the impact of diverse microenvironments on drug-combinations, here we develop a computational framework-Metabolism And GENomics-based Tailoring of Antibiotic regimens (MAGENTA). MAGENTA uses chemogenomic profiles of individual drugs and metabolic perturbations to predict synergistic or antagonistic drug-interactions in different microenvironments. We uncovered antibiotic combinations with robust synergy across nine distinct environments against both E. coli and A. baumannii by searching through 2556 drug-combinations of 72 drugs. MAGENTA also accurately predicted the change in efficacy of bacteriostatic and bactericidal drug-combinations during growth in glycerol media, which we confirmed experimentally in both microbes. Our approach identified genes in glycolysis and glyoxylate pathway as top predictors of synergy and antagonism respectively. Our systems approach enables tailoring of antibiotic therapies based on the pathogen microenvironment.


Asunto(s)
Antibacterianos/administración & dosificación , Modelos Biológicos , Pruebas de Farmacogenómica/métodos , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/crecimiento & desarrollo , Acinetobacter baumannii/metabolismo , Biología Computacional , Interacciones Farmacológicas , Farmacorresistencia Bacteriana Múltiple , Sinergismo Farmacológico , Quimioterapia Combinada , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Genes Bacterianos/genética , Glucólisis/efectos de los fármacos , Glucólisis/genética , Glioxilatos/metabolismo , Interacciones Microbiota-Huesped/efectos de los fármacos , Interacciones Microbiota-Huesped/genética , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Redes y Vías Metabólicas/genética , Pruebas de Sensibilidad Microbiana , Pruebas de Farmacogenómica/estadística & datos numéricos , Programas Informáticos , Biología de Sistemas
8.
Mol Syst Biol ; 12(5): 872, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27222539

RESUMEN

Combination antibiotic therapies are being increasingly used in the clinic to enhance potency and counter drug resistance. However, the large search space of candidate drugs and dosage regimes makes the identification of effective combinations highly challenging. Here, we present a computational approach called INDIGO, which uses chemogenomics data to predict antibiotic combinations that interact synergistically or antagonistically in inhibiting bacterial growth. INDIGO quantifies the influence of individual chemical-genetic interactions on synergy and antagonism and significantly outperforms existing approaches based on experimental evaluation of novel predictions in Escherichia coli Our analysis revealed a core set of genes and pathways (e.g. central metabolism) that are predictive of antibiotic interactions. By identifying the interactions that are associated with orthologous genes, we successfully estimated drug-interaction outcomes in the bacterial pathogens Mycobacterium tuberculosis and Staphylococcus aureus, using the E. coli INDIGO model. INDIGO thus enables the discovery of effective combination therapies in less-studied pathogens by leveraging chemogenomics data in model organisms.


Asunto(s)
Antibacterianos/farmacología , Biología Computacional/métodos , Escherichia coli/genética , Mycobacterium tuberculosis/genética , Staphylococcus aureus/genética , Bases de Datos de Compuestos Químicos , Bases de Datos Genéticas , Interacciones Farmacológicas , Quimioterapia Combinada , Escherichia coli/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos
9.
Mol Syst Biol ; 10: 740, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25028489

RESUMEN

Microbes can tailor transcriptional responses to diverse environmental challenges despite having streamlined genomes and a limited number of regulators. Here, we present data-driven models that capture the dynamic interplay of the environment and genome-encoded regulatory programs of two types of prokaryotes: Escherichia coli (a bacterium) and Halobacterium salinarum (an archaeon). The models reveal how the genome-wide distributions of cis-acting gene regulatory elements and the conditional influences of transcription factors at each of those elements encode programs for eliciting a wide array of environment-specific responses. We demonstrate how these programs partition transcriptional regulation of genes within regulons and operons to re-organize gene-gene functional associations in each environment. The models capture fitness-relevant co-regulation by different transcriptional control mechanisms acting across the entire genome, to define a generalized, system-level organizing principle for prokaryotic gene regulatory networks that goes well beyond existing paradigms of gene regulation. An online resource (http://egrin2.systemsbiology.net) has been developed to facilitate multiscale exploration of conditional gene regulation in the two prokaryotes.


Asunto(s)
Redes Reguladoras de Genes , Genoma Microbiano , Modelos Genéticos , Algoritmos , Escherichia coli/genética , Regulación de la Expresión Génica , Aptitud Genética , Halobacterium salinarum/genética , Operón , Elementos Reguladores de la Transcripción , Regulón
10.
PLoS Comput Biol ; 9(12): e1003370, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24348226

RESUMEN

There is a strong need for computational frameworks that integrate different biological processes and data-types to unravel cellular regulation. Current efforts to reconstruct transcriptional regulatory networks (TRNs) focus primarily on proximal data such as gene co-expression and transcription factor (TF) binding. While such approaches enable rapid reconstruction of TRNs, the overwhelming combinatorics of possible networks limits identification of mechanistic regulatory interactions. Utilizing growth phenotypes and systems-level constraints to inform regulatory network reconstruction is an unmet challenge. We present our approach Gene Expression and Metabolism Integrated for Network Inference (GEMINI) that links a compendium of candidate regulatory interactions with the metabolic network to predict their systems-level effect on growth phenotypes. We then compare predictions with experimental phenotype data to select phenotype-consistent regulatory interactions. GEMINI makes use of the observation that only a small fraction of regulatory network states are compatible with a viable metabolic network, and outputs a regulatory network that is simultaneously consistent with the input genome-scale metabolic network model, gene expression data, and TF knockout phenotypes. GEMINI preferentially recalls gold-standard interactions (p-value = 10(-172)), significantly better than using gene expression alone. We applied GEMINI to create an integrated metabolic-regulatory network model for Saccharomyces cerevisiae involving 25,000 regulatory interactions controlling 1597 metabolic reactions. The model quantitatively predicts TF knockout phenotypes in new conditions (p-value = 10(-14)) and revealed potential condition-specific regulatory mechanisms. Our results suggest that a metabolic constraint-based approach can be successfully used to help reconstruct TRNs from high-throughput data, and highlights the potential of using a biochemically-detailed mechanistic framework to integrate and reconcile inconsistencies across different data-types. The algorithm and associated data are available at https://sourceforge.net/projects/gemini-data/


Asunto(s)
Regulación de la Expresión Génica , Redes Reguladoras de Genes , Metabolismo , Transcripción Genética , Fenotipo , Levaduras/genética
11.
Proc Natl Acad Sci U S A ; 108(44): 18020-5, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21960440

RESUMEN

Using brain transcriptomic profiles from 853 individual honey bees exhibiting 48 distinct behavioral phenotypes in naturalistic contexts, we report that behavior-specific neurogenomic states can be inferred from the coordinated action of transcription factors (TFs) and their predicted target genes. Unsupervised hierarchical clustering of these transcriptomic profiles showed three clusters that correspond to three ecologically important behavioral categories: aggression, maturation, and foraging. To explore the genetic influences potentially regulating these behavior-specific neurogenomic states, we reconstructed a brain transcriptional regulatory network (TRN) model. This brain TRN quantitatively predicts with high accuracy gene expression changes of more than 2,000 genes involved in behavior, even for behavioral phenotypes on which it was not trained, suggesting that there is a core set of TFs that regulates behavior-specific gene expression in the bee brain, and other TFs more specific to particular categories. TFs playing key roles in the TRN include well-known regulators of neural and behavioral plasticity, e.g., Creb, as well as TFs better known in other biological contexts, e.g., NF-κB (immunity). Our results reveal three insights concerning the relationship between genes and behavior. First, distinct behaviors are subserved by distinct neurogenomic states in the brain. Second, the neurogenomic states underlying different behaviors rely upon both shared and distinct transcriptional modules. Third, despite the complexity of the brain, simple linear relationships between TFs and their putative target genes are a surprisingly prominent feature of the networks underlying behavior.


Asunto(s)
Conducta , Genómica , Transcripción Genética , Animales , Abejas/fisiología , Encéfalo/metabolismo
12.
PNAS Nexus ; 3(1): pgae013, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38292544

RESUMEN

Quiescence, a temporary withdrawal from the cell cycle, plays a key role in tissue homeostasis and regeneration. Quiescence is increasingly viewed as a continuum between shallow and deep quiescence, reflecting different potentials to proliferate. The depth of quiescence is altered in a range of diseases and during aging. Here, we leveraged genome-scale metabolic modeling (GEM) to define the metabolic and epigenetic changes that take place with quiescence deepening. We discovered contrasting changes in lipid catabolism and anabolism and diverging trends in histone methylation and acetylation. We then built a multi-cell type machine learning model that accurately predicts quiescence depth in diverse biological contexts. Using both machine learning and genome-scale flux simulations, we performed high-throughput screening of chemical and genetic modulators of quiescence and identified novel small molecule and genetic modulators with relevance to cancer and aging.

13.
STAR Protoc ; 5(3): 103173, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38970792

RESUMEN

Here, we present a protocol for analyzing the global metabolic landscape in breast tumors for the purpose of metabolism-based patient stratification. We describe steps for analyzing 1,454 metabolic genes representing 90 metabolic pathways and subjecting them to an algorithm that calculates the deregulation score of 90 pathways in each tumor sample, thus converting gene-level information into pathway-level information. We then detail procedures for performing clustering analysis to identify metabolic subtypes and using machine learning to develop a signature representing each subtype. For complete details on the use and execution of this protocol, please refer to Iqbal et al.1.

14.
iScience ; 27(2): 109025, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38357663

RESUMEN

Tuberculosis (TB) afflicted 10.6 million people in 2021, and its global burden is increasing due to multidrug-resistant TB (MDR-TB) and extensively resistant TB (XDR-TB). Here, we analyze multi-domain information from 5,060 TB patients spanning 10 countries with high burden of MDR-TB from the NIAID TB Portals database to determine predictors of TB treatment outcome. Our analysis revealed significant associations between radiological, microbiological, therapeutic, and demographic data modalities. Our machine learning model, built with 203 features across modalities outperforms models built using each modality alone in predicting treatment outcomes, with an accuracy of 83% and area under the curve of 0.84. Notably, our analysis revealed that the drug regimens Bedaquiline-Clofazimine-Cycloserine-Levofloxacin-Linezolid and Bedaquiline-Clofazimine-Linezolid-Moxifloxacin were associated with treatment success and failure, respectively, for MDR non-XDR-TB. Drug combinations predicted to be synergistic by the INDIGO algorithm performed better than antagonistic combinations. Our prioritized set of features predictive of treatment outcomes can ultimately guide the personalized clinical management of TB.

15.
bioRxiv ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38895385

RESUMEN

Machine learning (ML) algorithms are necessary to efficiently identify potent drug combinations within a large candidate space to combat drug resistance. However, existing ML approaches cannot be applied to emerging and under-studied pathogens with limited training data. To address this, we developed a transfer learning and crowdsourcing framework (TACTIC) to train ML models on data from multiple bacteria. TACTIC was built using 2,965 drug interactions from 12 bacterial strains and outperformed traditional ML models in predicting drug interaction outcomes for species that lack training data. Top TACTIC model features revealed genetic and metabolic factors that influence cross-species and species-specific drug interaction outcomes. Upon analyzing ~600,000 predicted drug interactions across 9 metabolic environments and 18 bacterial strains, we identified a small set of drug interactions that are selectively synergistic against Gram-negative (e.g., A. baumannii) and non-tuberculous mycobacteria (NTM) pathogens. We experimentally validated synergistic drug combinations containing clarithromycin, ampicillin, and mecillinam against M. abscessus, an emerging pathogen with growing levels of antibiotic resistance. Lastly, we leveraged TACTIC to propose selectively synergistic drug combinations to treat bacterial eye infections (endophthalmitis).

16.
Proc Natl Acad Sci U S A ; 107(41): 17845-50, 2010 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-20876091

RESUMEN

Prediction of metabolic changes that result from genetic or environmental perturbations has several important applications, including diagnosing metabolic disorders and discovering novel drug targets. A cardinal challenge in obtaining accurate predictions is the integration of transcriptional regulatory networks with the corresponding metabolic network. We propose a method called probabilistic regulation of metabolism (PROM) that achieves this synthesis and enables straightforward, automated, and quantitative integration of high-throughput data into constraint-based modeling, making it an ideal tool for constructing genome-scale regulatory-metabolic network models for less-studied organisms. PROM introduces probabilities to represent gene states and gene-transcription factor interactions. By using PROM, we constructed an integrated regulatory-metabolic network for the model organism, Escherichia coli, and demonstrated that our method based on automated inference is more accurate and comprehensive than the current state of the art, which is based on manual curation of literature. After validating the approach, we used PROM to build a genome-scale integrated metabolic-regulatory model for Mycobacterium tuberculosis, a critically important human pathogen. This study incorporated data from more than 1,300 microarrays, 2,000 transcription factor-target interactions regulating 3,300 metabolic reactions, and 1,905 KO phenotypes for E. coli and M. tuberculosis. PROM identified KO phenotypes with accuracies as high as 95%, and predicted growth rates quantitatively with correlation of 0.95. Importantly, PROM represents the successful integration of a top-down reconstructed, statistically inferred regulatory network with a bottom-up reconstructed, biochemically detailed metabolic network, bridging two important classes of systems biology models that are rarely combined quantitatively.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Escherichia coli/genética , Redes Reguladoras de Genes/genética , Redes y Vías Metabólicas/genética , Modelos Genéticos , Mycobacterium tuberculosis/genética , Biología de Sistemas/métodos , Genoma Bacteriano/genética
17.
Appl Biochem Biotechnol ; 195(4): 2648-2663, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35304691

RESUMEN

Environmental pollution is one of the major issues facing all countries throughout the world. Environmental degradation is occurring and creating crises in day-to-day life due to the increasing amount of chemicals used in industries, where even the effluents processed out after treatment also contain some trace elements. Hence the extraction of enzymes using natural methods is an alternative for the production of dye in order to reduce pollution, which in turn helps to nourish and protect the environment for future generations. Hibiscus sabdariffa (L.) is a rich source of anthocyanins that is further enhanced by callus formation and accumulated by increasing the sucrose concentration. Anthocyanin pigments were extracted using acidified ethanol. The dye obtained was screened by GC-MS analysis and its dyeing process used in the textile industry. The study showed certain properties affected the coloring nature depending on the cloth used. The color of anthocyanin pigment depends on the pH maintained and also shows adaptability to varied environmental conditions.


Asunto(s)
Antocianinas , Hibiscus , Antocianinas/química , Colorantes , Hibiscus/química , Extractos Vegetales/química , Textiles
18.
iScience ; 26(10): 108059, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37854701

RESUMEN

Extensive metabolic heterogeneity in breast cancers has limited the deployment of metabolic therapies. To enable patient stratification, we studied the metabolic landscape in breast cancers (∼3000 patients combined) and identified three subtypes with increasing degrees of metabolic deregulation. Subtype M1 was found to be dependent on bile-acid biosynthesis, whereas M2 showed reliance on methionine pathway, and M3 engaged fatty-acid, nucleotide, and glucose metabolism. The extent of metabolic alterations correlated strongly with tumor aggressiveness and patient outcome. This pattern was reproducible in independent datasets and using in vivo tumor metabolite data. Using machine-learning, we identified robust and generalizable signatures of metabolic subtypes in tumors and cell lines. Experimental inhibition of metabolic pathways in cell lines representing metabolic subtypes revealed subtype-specific sensitivity, therapeutically relevant drugs, and promising combination therapies. Taken together, metabolic stratification of breast cancers can thus aid in predicting patient outcome and designing precision therapies.

19.
PNAS Nexus ; 1(3): pgac132, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36016709

RESUMEN

Drug combinations are a promising strategy to counter antibiotic resistance. However, current experimental and computational approaches do not account for the entire complexity involved in combination therapy design, such as the effect of pathogen metabolic heterogeneity, changes in the growth environment, drug treatment order, and time interval. To address these limitations, we present a comprehensive approach that uses genome-scale metabolic modeling and machine learning to guide combination therapy design. Our mechanistic approach (a) accommodates diverse data types, (b) accounts for time- and order-specific interactions, and (c) accurately predicts drug interactions in various growth conditions and their robustness to pathogen metabolic heterogeneity. Our approach achieved high accuracy (area under the receiver operating curve (AUROC) = 0.83 for synergy, AUROC = 0.98 for antagonism) in predicting drug interactions for Escherichia coli cultured in 57 metabolic conditions based on experimental validation. The entropy in bacterial metabolic response was predictive of combination therapy outcomes across time scales and growth conditions. Simulation of metabolic heterogeneity using population FBA identified two subpopulations of E. coli cells defined by the levels of three proteins (eno, fadB, and fabD) in glycolysis and lipid metabolism that influence cell tolerance to a broad range of antibiotic combinations. Analysis of the vast landscape of condition-specific drug interactions revealed a set of 24 robustly synergistic drug combinations with potential for clinical use.

20.
Drug Discov Today ; 27(6): 1639-1651, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35398560

RESUMEN

Combination therapies can overcome antimicrobial resistance (AMR) and repurpose existing drugs. However, the large combinatorial space to explore presents a daunting challenge. In response, machine learning (ML) algorithms are being applied to identify novel synergistic drug interactions from millions of potential combinations. Here, we compare ML-based approaches for combination therapy design based on the type of input information used, specifically: drug properties, microbial response and infection microenvironment. We also provide a compilation of publicly available drug interaction datasets relevant to AMR. Finally, we discuss limitations of current ML-based methods and propose new strategies for designing efficacious combination therapies. These include consideration of in vivo conditions, design of sequential combinations, enhancement of model interpretability and application of deep learning algorithms.


Asunto(s)
Antiinfecciosos , Aprendizaje Automático , Algoritmos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/farmacología , Antiinfecciosos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA