Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(13): 2210-2212, 2022 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-35750032

RESUMO

Many approved drugs, including antivirals, are small-molecule inhibitors of disease-causing proteins. Such inhibitors often elicit resistance during treatment. Chaturvedi et al. propose new, feedback-disruptor (FD) antivirals that efficiently cure infected cells from viruses and minimize the chance of resistance, providing a new paradigm to treat viral infections and possibly other diseases.


Assuntos
Antivirais , Viroses , Antivirais/farmacologia , Antivirais/uso terapêutico , Retroalimentação , Humanos , Viroses/tratamento farmacológico
2.
Cell ; 144(6): 910-25, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21414483

RESUMO

Cellular decision making is the process whereby cells assume different, functionally important and heritable fates without an associated genetic or environmental difference. Such stochastic cell fate decisions generate nongenetic cellular diversity, which may be critical for metazoan development as well as optimized microbial resource utilization and survival in a fluctuating, frequently stressful environment. Here, we review several examples of cellular decision making from viruses, bacteria, yeast, lower metazoans, and mammals, highlighting the role of regulatory network structure and molecular noise. We propose that cellular decision making is one of at least three key processes underlying development at various scales of biological organization.


Assuntos
Diferenciação Celular , Modelos Biológicos , Animais , Humanos
3.
Proc Natl Acad Sci U S A ; 120(49): e2303114120, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38019857

RESUMO

Drug resistance continues to impede the success of cancer treatments, creating a need for experimental model systems that are broad, yet simple, to allow the identification of mechanisms and novel countermeasures applicable to many cancer types. To address these needs, we investigated a set of engineered mammalian cell lines with synthetic gene circuits integrated into their genome that evolved resistance to Puromycin. We identified DNA amplification as the mechanism underlying drug resistance in 4 out of 6 replicate populations. Triplex-forming oligonucleotide (TFO) treatment combined with Puromycin could efficiently suppress the growth of cell populations with DNA amplification. Similar observations in human cancer cell lines suggest that TFOs could be broadly applicable to mitigate drug resistance, one of the major difficulties in treating cancer.


Assuntos
DNA , Neoplasias , Animais , Humanos , DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Genes Sintéticos , Oligonucleotídeos , Puromicina , Mamíferos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética
4.
Nat Chem Biol ; 19(7): 887-899, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37231268

RESUMO

A major pharmacological assumption is that lowering disease-promoting protein levels is generally beneficial. For example, inhibiting metastasis activator BACH1 is proposed to decrease cancer metastases. Testing such assumptions requires approaches to measure disease phenotypes while precisely adjusting disease-promoting protein levels. Here we developed a two-step strategy to integrate protein-level tuning, noise-aware synthetic gene circuits into a well-defined human genomic safe harbor locus. Unexpectedly, engineered MDA-MB-231 metastatic human breast cancer cells become more, then less and then more invasive as we tune BACH1 levels up, irrespective of the native BACH1. BACH1 expression shifts in invading cells, and expression of BACH1's transcriptional targets confirm BACH1's nonmonotone phenotypic and regulatory effects. Thus, chemical inhibition of BACH1 could have unwanted effects on invasion. Additionally, BACH1's expression variability aids invasion at high BACH1 expression. Overall, precisely engineered, noise-aware protein-level control is necessary and important to unravel disease effects of genes to improve clinical drug efficacy.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica , Neoplasias da Mama , Humanos , Feminino , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Neoplasias da Mama/metabolismo , Metástase Neoplásica
5.
Biophys J ; 122(13): 2623-2635, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37218129

RESUMO

Gene expression is inherently noisy due to small numbers of proteins and nucleic acids inside a cell. Likewise, cell division is stochastic, particularly when tracking at the level of a single cell. The two can be coupled when gene expression affects the rate of cell division. Single-cell time-lapse experiments can measure both fluctuations by simultaneously recording protein levels inside a cell and its stochastic division. These information-rich noisy trajectory data sets can be harnessed to learn about the underlying molecular and cellular details that are often not known a priori. A critical question is: How can we infer a model given data where fluctuations at two levels-gene expression and cell division-are intricately convoluted? We show the principle of maximum caliber (MaxCal)-integrated within a Bayesian framework-can be used to infer several cellular and molecular details (division rates, protein production, and degradation rates) from these coupled stochastic trajectories (CSTs). We demonstrate this proof of concept using synthetic data generated from a known model. An additional challenge in data analysis is that trajectories are often not in protein numbers, but in noisy fluorescence that depends on protein number in a probabilistic manner. We again show that MaxCal can infer important molecular and cellular rates even when data are in fluorescence, another example of CST with three confounding factors-gene expression noise, cell division noise, and fluorescence distortion-all coupled. Our approach will provide guidance to build models in synthetic biology experiments as well as general biological systems where examples of CSTs are abundant.


Assuntos
Modelos Biológicos , Proteínas , Teorema de Bayes , Divisão Celular , Proteínas/metabolismo , Expressão Gênica , Processos Estocásticos
6.
J Theor Biol ; 575: 111630, 2023 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-37804940

RESUMO

Understanding the potential for cancers to metastasize is still relatively unknown. While many predictive methods may use deep learning or stochastic processes, we highlight a long standing mathematical concept that may be useful for modeling metastatic breast cancer systems. Ordinary differential equations (ODEs) can model cell state transitions by considering the pertinent environmental variables as well as the paths systems take over time. Bifurcation theory is a branch of dynamical systems which studies changes in the behavior of an ODE system while one or more parameters are varied. Many studies have applied concepts in one-parameter bifurcation theory to model biological network dynamics, and cell division. However, studies of two-parameter bifurcations are much more rare. Two-parameter bifurcations have not been studied in metastatic systems. Here we show how a specific two-parameter bifurcation phenomenon called a cusp bifurcation separates two qualitatively different metastatic cell state transitions modalities and propose a new perspective on defining such transitions based on mathematical theory. We hope the observations and verification methods detailed here may help in the understanding of metastatic potential from a basic biological perspective and in clinical settings.


Assuntos
Conceitos Matemáticos , Modelos Biológicos , Processos Estocásticos , Tempo , Divisão Celular
7.
Proc Natl Acad Sci U S A ; 116(50): 25162-25171, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31754027

RESUMO

Evolutionary reversibility-the ability to regain a lost function-is an important problem both in evolutionary and synthetic biology, where repairing natural or synthetic systems broken by evolutionary processes may be valuable. Here, we use a synthetic positive-feedback (PF) gene circuit integrated into haploid Saccharomyces cerevisiae cells to test if the population can restore lost PF function. In previous evolution experiments, mutations in a gene eliminated the fitness costs of PF activation. Since PF activation also provides drug resistance, exposing such compromised or broken mutants to both drug and inducer should create selection pressure to regain drug resistance and possibly PF function. Indeed, evolving 7 PF mutant strains in the presence of drug revealed 3 adaptation scenarios through genomic, PF-external mutations that elevate PF basal expression, possibly by affecting transcription, translation, degradation, and other fundamental cellular processes. Nonfunctional mutants gained drug resistance without ever developing high expression, while quasifunctional and dysfunctional PF mutants developed high expression nongenetically, which then diminished, although more slowly for dysfunctional mutants where revertant clones arose. These results highlight how intracellular context, such as the growth rate, can affect regulatory network dynamics and evolutionary dynamics, which has important consequences for understanding the evolution of drug resistance and developing future synthetic biology applications.


Assuntos
Evolução Molecular , Redes Reguladoras de Genes , Saccharomyces cerevisiae/genética , Aptidão Genética , Mutação , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Biologia Sintética
8.
Trends Genet ; 34(10): 733-735, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30119990

RESUMO

The correct expression of genes is vital for cells to function. Schikora-Tamarit et al. show that, in addition to obeying their promoters, most genes can modulate their own expression by either buffering or amplification. This could help to avoid costly overexpression of proteins.


Assuntos
Fatores de Necrose Tumoral , Citocina TWEAK , Regiões Promotoras Genéticas
9.
Nucleic Acids Res ; 47(14): 7703-7714, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31269201

RESUMO

Gene autorepression is widely present in nature and is also employed in synthetic biology, partly to reduce gene expression noise in cells. Optogenetic systems have recently been developed for controlling gene expression levels in mammalian cells, but most have utilized activator-based proteins, neglecting negative feedback except for in silico control. Here, we engineer optogenetic gene circuits into mammalian cells to achieve noise-reduction for precise gene expression control by genetic, in vitro negative feedback. We build a toolset of these noise-reducing Light-Inducible Tuner (LITer) gene circuits using the TetR repressor fused with a Tet-inhibiting peptide (TIP) or a degradation tag through the light-sensitive LOV2 protein domain. These LITers provide a range of nearly 4-fold gene expression control and up to 5-fold noise reduction from existing optogenetic systems. Moreover, we use the LITer gene circuit architecture to control gene expression of the cancer oncogene KRAS(G12V) and study its downstream effects through phospho-ERK levels and cellular proliferation. Overall, these novel LITer optogenetic platforms should enable precise spatiotemporal perturbations for studying multicellular phenotypes in developmental biology, oncology and other biomedical fields of research.


Assuntos
Retroalimentação Fisiológica , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Optogenética/métodos , Algoritmos , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos da radiação , Redes Reguladoras de Genes/efeitos da radiação , Células HEK293 , Humanos , Luz , Modelos Genéticos , Proteínas Proto-Oncogênicas p21(ras)/genética , Reprodutibilidade dos Testes , Biologia Sintética/métodos
10.
Proc Natl Acad Sci U S A ; 115(45): E10797-E10806, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30341217

RESUMO

Most organisms must cope with temperature changes. This involves genes and gene networks both as subjects and agents of cellular protection, creating difficulties in understanding. Here, we study how heating and cooling affect expression of single genes and synthetic gene circuits in Saccharomyces cerevisiae We discovered that nonoptimal temperatures induce a cell fate choice between stress resistance and growth arrest. This creates dramatic gene expression bimodality in isogenic cell populations, as arrest abolishes gene expression. Multiscale models incorporating population dynamics, temperature-dependent growth rates, and Arrhenius scaling of reaction rates captured the effects of cooling, but not those of heating in resistant cells. Molecular-dynamics simulations revealed how heating alters the conformational dynamics of the TetR repressor, fully explaining the experimental observations. Overall, nonoptimal temperatures induce a cell fate decision and corrupt gene and gene network function in computationally predictable ways, which may aid future applications of engineered microbes in nonstandard temperatures.


Assuntos
Adaptação Fisiológica/genética , Pontos de Checagem do Ciclo Celular/genética , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Redes Reguladoras de Genes , Saccharomyces cerevisiae/genética , Temperatura Baixa , Proteínas Fúngicas/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Temperatura Alta , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Saccharomyces cerevisiae/metabolismo , Estresse Fisiológico , Termodinâmica
11.
PLoS Biol ; 15(5): e2000644, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28486496

RESUMO

Genetically identical cells frequently display substantial heterogeneity in gene expression, cellular morphology and physiology. It has been suggested that by rapidly generating a subpopulation with novel phenotypic traits, phenotypic heterogeneity (or plasticity) accelerates the rate of adaptive evolution in populations facing extreme environmental challenges. This issue is important as cell-to-cell phenotypic heterogeneity may initiate key steps in microbial evolution of drug resistance and cancer progression. Here, we study how stochastic transitions between cellular states influence evolutionary adaptation to a stressful environment in yeast Saccharomyces cerevisiae. We developed inducible synthetic gene circuits that generate varying degrees of expression stochasticity of an antifungal resistance gene. We initiated laboratory evolutionary experiments with genotypes carrying different versions of the genetic circuit by exposing the corresponding populations to gradually increasing antifungal stress. Phenotypic heterogeneity altered the evolutionary dynamics by transforming the adaptive landscape that relates genotype to fitness. Specifically, it enhanced the adaptive value of beneficial mutations through synergism between cell-to-cell variability and genetic variation. Our work demonstrates that phenotypic heterogeneity is an evolving trait when populations face a chronic selection pressure. It shapes evolutionary trajectories at the genomic level and facilitates evolutionary rescue from a deteriorating environmental stress.


Assuntos
Adaptação Biológica , Evolução Biológica , Farmacorresistência Fúngica/genética , Genes Fúngicos , Fenótipo , Mutação , Saccharomyces cerevisiae
13.
Entropy (Basel) ; 22(4)2020 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-33286254

RESUMO

The promoter state of a gene and its expression levels are modulated by the amounts of transcription factors interacting with its regulatory regions. Hence, one may interpret a gene network as a communicating system in which the state of the promoter of a gene (the source) is communicated by the amounts of transcription factors that it expresses (the message) to modulate the state of the promoter and expression levels of another gene (the receptor). The reliability of the gene network dynamics can be quantified by Shannon's entropy of the message and the mutual information between the message and the promoter state. Here we consider a stochastic model for a binary gene and use its exact steady state solutions to calculate the entropy and mutual information. We show that a slow switching promoter with long and equally standing ON and OFF states maximizes the mutual information and reduces entropy. That is a binary gene expression regime generating a high variance message governed by a bimodal probability distribution with peaks of the same height. Our results indicate that Shannon's theory can be a powerful framework for understanding how bursty gene expression conciliates with the striking spatio-temporal precision exhibited in pattern formation of developing organisms.

14.
PLoS Pathog ; 13(5): e1006355, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28489916

RESUMO

Similar to other yeasts, the human pathogen Candida glabrata ages when it undergoes asymmetric, finite cell divisions, which determines its replicative lifespan. We sought to investigate if and how aging changes resilience of C. glabrata populations in the host environment. Our data demonstrate that old C. glabrata are more resistant to hydrogen peroxide and neutrophil killing, whereas young cells adhere better to epithelial cell layers. Consequently, virulence of old compared to younger C. glabrata cells is enhanced in the Galleria mellonella infection model. Electron microscopy images of old C. glabrata cells indicate a marked increase in cell wall thickness. Comparison of transcriptomes of old and young C. glabrata cells reveals differential regulation of ergosterol and Hog pathway associated genes as well as adhesion proteins, and suggests that aging is accompanied by remodeling of the fungal cell wall. Biochemical analysis supports this conclusion as older cells exhibit a qualitatively different lipid composition, leading to the observed increased emergence of fluconazole resistance when grown in the presence of fluconazole selection pressure. Older C. glabrata cells accumulate during murine and human infection, which is statistically unlikely without very strong selection. Therefore, we tested the hypothesis that neutrophils constitute the predominant selection pressure in vivo. When we altered experimentally the selection pressure by antibody-mediated removal of neutrophils, we observed a significantly younger pathogen population in mice. Mathematical modeling confirmed that differential selection of older cells is sufficient to cause the observed demographic shift in the fungal population. Hence our data support the concept that pathogenesis is affected by the generational age distribution of the infecting C. glabrata population in a host. We conclude that replicative aging constitutes an emerging trait, which is selected by the host and may even play an unanticipated role in the transition from a commensal to a pathogen state.


Assuntos
Candida glabrata/fisiologia , Candida glabrata/patogenicidade , Candidíase/microbiologia , Farmacorresistência Fúngica , Animais , Antifúngicos/farmacologia , Candida glabrata/efeitos dos fármacos , Candida glabrata/genética , Adesão Celular , Divisão Celular , Parede Celular/ultraestrutura , Fluconazol/farmacologia , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Camundongos , Mariposas , Neutrófilos/microbiologia , Fenótipo , Seleção Genética , Análise de Sequência de RNA , Fatores de Tempo , Virulência
15.
In Silico Biol ; 13(1-2): 21-39, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30562900

RESUMO

 Quantitative modeling is quickly becoming an integral part of biology, due to the ability of mathematical models and computer simulations to generate insights and predict the behavior of living systems. Single-cell models can be incapable or misleading for inferring population dynamics, as they do not consider the interactions between cells via metabolites or physical contact, nor do they consider competition for limited resources such as nutrients or space. Here we examine methods that are commonly used to model and simulate cell populations. First, we cover simple models where analytic solutions are available, and then move on to more complex scenarios where computational methods are required. Overall, we present a summary of mathematical models used to describe cell population dynamics, which may aid future model development and highlights the importance of population modeling in biology.


Assuntos
Fenômenos Fisiológicos Celulares , Modelos Biológicos , Algoritmos , Diferenciação Celular , Divisão Celular , Proliferação de Células , Cadeias de Markov , Método de Monte Carlo
16.
Phys Biol ; 16(3): 031002, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30654341

RESUMO

We present the epithelial-to-mesenchymal transition (EMT) from two perspectives: experimental/technological and theoretical. We review the state of the current understanding of the regulatory networks that underlie EMT in three physiological contexts: embryonic development, wound healing, and metastasis. We describe the existing experimental systems and manipulations used to better understand the molecular participants and factors that influence EMT and metastasis. We review the mathematical models of the regulatory networks involved in EMT, with a particular emphasis on the network motifs (such as coupled feedback loops) that can generate intermediate hybrid states between the epithelial and mesenchymal states. Ultimately, the understanding gained about these networks should be translated into methods to control phenotypic outcomes, especially in the context of cancer therapeutic strategies. We present emerging theories of how to drive the dynamics of a network toward a desired dynamical attractor (e.g. an epithelial cell state) and emerging synthetic biology technologies to monitor and control the state of cells.


Assuntos
Desenvolvimento Embrionário/fisiologia , Transição Epitelial-Mesenquimal , Metástase Neoplásica/fisiopatologia , Cicatrização/fisiologia , Desenvolvimento Embrionário/genética , Redes Reguladoras de Genes , Modelos Teóricos , Metástase Neoplásica/genética , Cicatrização/genética
17.
J Theor Biol ; 483: 110005, 2019 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-31525321

RESUMO

Lysogens are bacterial cells that have survived after genomically incorporating the DNA of temperate bacteriophages infecting them. If an infection results in lysogeny, the lysogen continues to grow and divide normally, seemingly unaffected by the integrated viral genome known as a prophage. However, the prophage can still have an impact on the host's phenotype and overall fitness in certain environments. Additionally, the prophage within the lysogen can activate the lytic pathway via spontaneous prophage induction (SPI), killing the lysogen and releasing new progeny phages. These new phages can then lyse or lysogenize other susceptible nonlysogens, thereby impacting the competition between lysogens and nonlysogens. In a scenario with differing growth rates, it is not clear whether SPI would be beneficial or detrimental to the lysogens since it kills the host cell but also attacks nonlysogenic competitors, either lysing or lysogenizing them. Here we study the evolutionary dynamics of a mixture of lysogens and nonlysogens and derive general conditions on SPI rates for lysogens to displace nonlysogens. We show that there exists an optimal SPI rate for bacteriophage λ and explain why it is so low. We also investigate the impact of stochasticity and conclude that even at low cell numbers SPI can still provide an advantage to the lysogens. These results corroborate recent experimental studies showing that lower SPI rates are advantageous for phage-phage competition, and establish theoretical bounds on the SPI rate in terms of ecological and environmental variables associated with lysogens having a competitive advantage over their nonlysogenic counterparts.


Assuntos
Prófagos/fisiologia , Simulação por Computador , Lisogenia , Modelos Biológicos , Probabilidade , Processos Estocásticos
18.
Biophys J ; 113(9): 2121-2130, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29117534

RESUMO

Learning the underlying details of a gene network is a major challenge in cellular and synthetic biology. We address this challenge by building a chemical kinetic model that utilizes information encoded in the stochastic protein expression trajectories typically measured in experiments. The applicability of the proposed method is demonstrated in an auto-activating genetic circuit, a common motif in natural and synthetic gene networks. Our approach is based on the principle of maximum caliber (MaxCal)-a dynamical analog of the principle of maximum entropy-and builds a minimal model using only three constraints: 1) protein synthesis, 2) protein degradation, and 3) positive feedback. The MaxCal-generated model (described with four parameters) was benchmarked against synthetic data generated using a Gillespie algorithm on a known reaction network (with seven parameters). MaxCal accurately predicts underlying rate parameters of protein synthesis and degradation as well as experimental observables such as protein number and dwell-time distributions. Furthermore, MaxCal yields an effective feedback parameter that can be useful for circuit design. We also extend our methodology and demonstrate how to analyze trajectories that are not in protein numbers but in arbitrary fluorescence units, a more typical condition in experiments. This "top-down" methodology based on minimal information-in contrast to traditional "bottom-up" approaches that require ad hoc knowledge of circuit details-provides a powerful tool to accurately infer underlying details of feedback circuits that are not otherwise visible in experiments and to help guide circuit design.


Assuntos
Redes Reguladoras de Genes , Modelos Genéticos , Funções Verossimilhança
19.
Biophys J ; 113(9): 2110-2120, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29117533

RESUMO

Gene regulatory networks are largely responsible for cellular decision-making. These networks sense diverse external signals and respond by adjusting gene expression, enabling cells to reach environment-dependent decisions crucial for their survival or reproduction. However, information-carrying signals may arrive at variable times. Besides the intrinsic strength of these signals, their arrival time (timing) may also carry information about the environment and can influence cellular decision-making in ways that are poorly understood. For example, it is unclear how the timing of individual phage infections affects the lysis-lysogeny decision of bacteriophage λ despite variable infection times being likely in the wild and even in laboratory conditions. In this work, we combine mathematical modeling with experimentation to address this question. We develop an experimentally testable theory, which reveals that late-infecting phages contribute less to cellular decision-making. This implies that infection delays lower the probability of lysogeny compared to simultaneous infections. Furthermore, we show that infection delays reduce lysogenization by providing insufficient CII for threshold crossing during the critical decision-making period. We find evidence for a cutoff time after which subsequent infections cannot influence the cellular decision. We derive an intuitive formula that approximates the probability of lysogeny for variable infection times by a time-weighted average of probabilities for simultaneous infections. We validate these theoretical predictions experimentally. Similar concepts and simplifying modeling approaches may help elucidate the mechanisms underlying other cellular decisions.


Assuntos
Lisogenia , Modelos Biológicos , Transdução de Sinais , Bacteriófago lambda/fisiologia , Escherichia coli/citologia , Escherichia coli/genética , Escherichia coli/fisiologia , Escherichia coli/virologia , Redes Reguladoras de Genes , Processos Estocásticos
20.
PLoS Comput Biol ; 12(3): e1004825, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27010473

RESUMO

Metabolic efficiency depends on the balance between supply and demand of metabolites, which is sensitive to environmental and physiological fluctuations, or noise, causing shortages or surpluses in the metabolic pipeline. How cells can reliably optimize biomass production in the presence of metabolic fluctuations is a fundamental question that has not been fully answered. Here we use mathematical models to predict that enzyme saturation creates distinct regimes of cellular growth, including a phase of growth arrest resulting from toxicity of the metabolic process. Noise can drive entry of single cells into growth arrest while a fast-growing majority sustains the population. We confirmed these predictions by measuring the growth dynamics of Escherichia coli utilizing lactose as a sole carbon source. The predicted heterogeneous growth emerged at high lactose concentrations, and was associated with cell death and production of antibiotic-tolerant persister cells. These results suggest how metabolic networks may balance costs and benefits, with important implications for drug tolerance.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Enzimas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/citologia , Escherichia coli/crescimento & desenvolvimento , Modelos Biológicos , Proliferação de Células/fisiologia , Simulação por Computador , Ativação Enzimática , Taxa de Depuração Metabólica , Proteínas de Transporte de Monossacarídeos/metabolismo , Simportadores/metabolismo , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa