Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Methods Mol Biol ; 2813: 167-188, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38888778

RESUMEN

Quantification of Mycobacterium tuberculosis (Mtb) growth dynamics in cell-based in vitro infection models is traditionally carried out by measurement of colony forming units (CFU). However, Mtb being an extremely slow growing organism (16-24 h doubling time), this approach requires at least 3 weeks of incubation to obtain measurable readouts. In this chapter, we describe an alternative approach based on time-lapse microscopy and quantitative image analysis that allows faster quantification of Mtb growth dynamics in host cells. In addition, this approach provides the capability to capture other readouts from the same experimental setup, such as host cell viability, bacterial localization as well as the dynamics of propagation of infection between the host cells.


Asunto(s)
Microscopía Fluorescente , Mycobacterium tuberculosis , Imagen de Lapso de Tiempo , Mycobacterium tuberculosis/crecimiento & desarrollo , Imagen de Lapso de Tiempo/métodos , Microscopía Fluorescente/métodos , Humanos , Tuberculosis/microbiología , Procesamiento de Imagen Asistido por Computador/métodos , Interacciones Huésped-Patógeno
2.
Nat Commun ; 15(1): 1550, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38378733

RESUMEN

Super-resolution techniques expand the abilities of researchers who have the knowledge and resources to either build or purchase a system. This excludes the part of the research community without these capabilities. Here we introduce the openSIM add-on to upgrade existing optical microscopes to Structured Illumination super-resolution Microscopes (SIM). The openSIM is an open-hardware system, designed and documented to be easily duplicated by other laboratories, making super-resolution modality accessible to facilitate innovative research. The add-on approach gives a performance improvement for pre-existing lab equipment without the need to build a completely new system.

3.
Sci Adv ; 10(1): eadh7957, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38170768

RESUMEN

Invading microbes face a myriad of cidal mechanisms of phagocytes that inflict physical damage to microbial structures. How intracellular bacterial pathogens adapt to these stresses is not fully understood. Here, we report the discovery of a virulence mechanism by which changes to the mechanical stiffness of the mycobacterial cell surface confer refraction to killing during infection. Long-term time-lapse atomic force microscopy was used to reveal a process of "mechanical morphotype switching" in mycobacteria exposed to host intracellular stress. A "soft" mechanical morphotype switch enhances tolerance to intracellular macrophage stress, including cathelicidin. Both pharmacologic treatment, with bedaquiline, and a genetic mutant lacking uvrA modified the basal mechanical state of mycobacteria into a soft mechanical morphotype, enhancing survival in macrophages. Our study proposes microbial cell mechanical adaptation as a critical axis for surviving host-mediated stressors.


Asunto(s)
Mycobacterium , Macrófagos/metabolismo , Fagocitos , Membrana Celular
5.
Cell ; 186(23): 5135-5150.e28, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37865090

RESUMEN

Mycobacterium tuberculosis (Mtb) cultured axenically without detergent forms biofilm-like cords, a clinical identifier of virulence. In lung-on-chip (LoC) and mouse models, cords in alveolar cells contribute to suppression of innate immune signaling via nuclear compression. Thereafter, extracellular cords cause contact-dependent phagocyte death but grow intercellularly between epithelial cells. The absence of these mechanopathological mechanisms explains the greater proportion of alveolar lesions with increased immune infiltration and dissemination defects in cording-deficient Mtb infections. Compression of Mtb lipid monolayers induces a phase transition that enables mechanical energy storage. Agent-based simulations demonstrate that the increased energy storage capacity is sufficient for the formation of cords that maintain structural integrity despite mechanical perturbation. Bacteria in cords remain translationally active despite antibiotic exposure and regrow rapidly upon cessation of treatment. This study provides a conceptual framework for the biophysics and function in tuberculosis infection and therapy of cord architectures independent of mechanisms ascribed to single bacteria.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Ratones , Biopelículas , Pulmón/microbiología , Pulmón/patología , Mycobacterium tuberculosis/fisiología , Tuberculosis/microbiología , Tuberculosis/patología , Virulencia , Fenómenos Biomecánicos
6.
Elife ; 122023 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-37530405

RESUMEN

A DNA damage-inducible mutagenic gene cassette has been implicated in the emergence of drug resistance in Mycobacterium tuberculosis during anti-tuberculosis (TB) chemotherapy. However, the molecular composition and operation of the encoded 'mycobacterial mutasome' - minimally comprising DnaE2 polymerase and ImuA' and ImuB accessory proteins - remain elusive. Following exposure of mycobacteria to DNA damaging agents, we observe that DnaE2 and ImuB co-localize with the DNA polymerase III ß subunit (ß clamp) in distinct intracellular foci. Notably, genetic inactivation of the mutasome in an imuBAAAAGG mutant containing a disrupted ß clamp-binding motif abolishes ImuB-ß clamp focus formation, a phenotype recapitulated pharmacologically by treating bacilli with griselimycin and in biochemical assays in which this ß clamp-binding antibiotic collapses pre-formed ImuB-ß clamp complexes. These observations establish the essentiality of the ImuB-ß clamp interaction for mutagenic DNA repair in mycobacteria, identifying the mutasome as target for adjunctive therapeutics designed to protect anti-TB drugs against emerging resistance.


Asunto(s)
Proteínas Bacterianas , Mycobacterium tuberculosis , Proteínas Bacterianas/química , Mycobacterium tuberculosis/genética , Mutagénesis , Reparación del ADN , Antituberculosos/farmacología
7.
EMBO J ; 42(9): e113490, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36920246

RESUMEN

Mycobacterium tuberculosis (Mtb) infection is initiated by inhalation of bacteria into lung alveoli, where they are phagocytosed by resident macrophages. Intracellular Mtb replication induces the death of the infected macrophages and the release of bacterial aggregates. Here, we show that these aggregates can evade phagocytosis by killing macrophages in a contact-dependent but uptake-independent manner. We use time-lapse fluorescence microscopy to show that contact with extracellular Mtb aggregates triggers macrophage plasma membrane perturbation, cytosolic calcium accumulation, and pyroptotic cell death. These effects depend on the Mtb ESX-1 secretion system, however, this system alone cannot induce calcium accumulation and macrophage death in the absence of the Mtb surface-exposed lipid phthiocerol dimycocerosate. Unexpectedly, we found that blocking ESX-1-mediated secretion of the EsxA/EsxB virulence factors does not eliminate the uptake-independent killing of macrophages and that the 50-kDa isoform of the ESX-1-secreted protein EspB can mediate killing in the absence of EsxA/EsxB secretion. Treatment with an ESX-1 inhibitor reduces uptake-independent killing of macrophages by Mtb aggregates, suggesting that novel therapies targeting this anti-phagocytic mechanism could prevent the propagation of extracellular bacteria within the lung.


Asunto(s)
Mycobacterium tuberculosis , Proteínas Bacterianas/metabolismo , Calcio/metabolismo , Macrófagos/metabolismo , Factores de Virulencia/metabolismo
8.
mBio ; 13(5): e0225122, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36121153

RESUMEN

Mycobacterium tuberculosis infection is initiated by the inhalation and implantation of bacteria in the lung alveoli, where they are phagocytosed by macrophages. Even a single bacterium may be sufficient to initiate infection. Thereafter, the clinical outcome is highly variable between individuals, ranging from sterilization to active disease, for reasons that are not well understood. Here, we show that the rate of intracellular bacterial growth varies markedly between individual macrophages, and this heterogeneity is driven by cell-to-cell variation of inducible nitric oxide synthase (iNOS) activity. At the single-cell level, iNOS expression fluctuates over time, independent of infection or activation with gamma interferon. We conclude that chance encounters between individual bacteria and host cells randomly expressing different levels of an antibacterial gene can determine the outcome of single-cell infections, which may explain why some exposed individuals clear the bacteria while others develop progressive disease. IMPORTANCE In this report, we demonstrate that fluctuations in the expression of antimicrobial genes can define how single host cells control bacterial infections. We show that preexisting cell-to-cell variation in the expression of a single gene, that for inducible nitric oxide synthase, is sufficient to explain why some macrophages kill intracellular M. tuberculosis while others fail to control bacterial replication, possibly leading to disease progression. We introduce the concept that chance encounters between heterogeneous bacteria and host cells can determine the outcome of a host-pathogen interaction. This concept is particularly relevant for all the infectious diseases in which the number of interacting pathogens and host cells is small at some point during the infection.


Asunto(s)
Mycobacterium tuberculosis , Humanos , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Mycobacterium tuberculosis/metabolismo , Interferón gamma/metabolismo , Óxido Nítrico Sintasa/genética , Macrófagos/microbiología , Antibacterianos/metabolismo , Óxido Nítrico/metabolismo
9.
Oncotarget ; 12(20): 2075-2088, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34611481

RESUMEN

PURPOSE: To investigate safety, response, and survival after ablative glass microsphere 90Y radioembolization for unresectable intrahepatic cholangiocarcinoma. MATERIALS AND METHODS: A retrospective review of 37 radioembolizations in 28 patients treated with single compartment dose of ≥190 Gy encompassing >75% of the largest tumor was performed. Tumors were assessed for stage, morphology, and arterial supply. Response per Modified Response Evaluation Criteria in Solid Tumors (mRECIST), freedom from progression (FFP), progression-free survival (PFS), overall survival (OS), biochemical hepatic function, performance status, and adverse events were investigated. RESULTS: The median highest dose per patient was 256.8 Gy (195.7-807.8). Objective response at 3 months was 94.1% (complete 44.1% and partial 50%). Median OS was not reached and the 30-month OS rate was 59%, with a median follow-up of 13.4 months (5.4-39.4). FFP in the radiated field and overall FFP at 30 months were 67% and 40%, respectively. Favorable arterial supply was associated with improved OS (p = 0.018). Unfavorable arterial supply was associated with worse OS [HR 5.7 (95% CI 1.1-28.9, p = 0.034)], and PFS [HR 5.9 (95% CI 1.9-18.4, p = 0.002)]. Patients with mass-forming tumors had a survival benefit (p = 0.002). Laboratory values and performance status did not significantly change 3 months after radioembolization. Grade 3 and 4 adverse events occurred in 2 (7.1%) patients. CONCLUSIONS: Radioembolization of unresectable intrahepatic cholangiocarcinoma with ablative intent has a high response rate, promising survival, and is well tolerated.

10.
Elife ; 102021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34219648

RESUMEN

Uropathogenic Escherichia coli (UPEC) proliferate within superficial bladder umbrella cells to form intracellular bacterial communities (IBCs) during early stages of urinary tract infections. However, the dynamic responses of IBCs to host stresses and antibiotic therapy are difficult to assess in situ. We develop a human bladder-chip model wherein umbrella cells and bladder microvascular endothelial cells are co-cultured under flow in urine and nutritive media respectively, and bladder filling and voiding mimicked mechanically by application and release of linear strain. Using time-lapse microscopy, we show that rapid recruitment of neutrophils from the vascular channel to sites of infection leads to swarm and neutrophil extracellular trap formation but does not prevent IBC formation. Subsequently, we tracked bacterial growth dynamics in individual IBCs through two cycles of antibiotic administration interspersed with recovery periods which revealed that the elimination of bacteria within IBCs by the antibiotic was delayed, and in some instances, did not occur at all. During the recovery period, rapid proliferation in a significant fraction of IBCs reseeded new foci of infection through bacterial shedding and host cell exfoliation. These insights reinforce a dynamic role for IBCs as harbors of bacterial persistence, with significant consequences for non-compliance with antibiotic regimens.


Urinary tract infections are one of the most common reasons people need antibiotics. These bacterial infections are typically caused by uropathogenic Escherichia coli (also known as UPEC), which either float freely in the urine and wash away when the bladder empties, or form communities inside cells that the bladder struggles to clear. It is possible that the bacteria living within cells are also more protected from the immune system and antibiotics. But this is hard to study in animal models. To overcome this, Sharma et al. built a 'bladder-chip' which mimics the interface between the blood vessels and the tissue layers of the human bladder. Similar chip devices have also been made for other organs. However, until now, no such model had been developed for the bladder. On the chip created by Sharma et al. is a layer of bladder cells which sit at the bottom of a channel filled with diluted human urine. These cells were infected with UPEC, and then imaged over time to see how the bacteria moved, interacted with the bladder cells, and aggregated together. Immune cells from human blood were then added to a vascular channel underneath the bladder tissue, which is coated with endothelial cells that normally line blood vessels. The immune cells rapidly crossed the endothelial barrier and entered the bladder tissue, and swarmed around sites of infection. In some instances, they released the contents of their cells to form net-like traps to catch the bacteria. But these traps failed to remove the bacteria living inside bladder cells. Antibiotics were then added to the urine flowing over the bladder cells as well as the vascular channel, similar to how drugs would be delivered in live human tissue. Sharma et al. discovered that the antibiotics killed bacteria residing in bladder cells slower than bacteria floating freely in the urine. Furthermore, they found that bacteria living in tightly packed communities within bladder cells were more likely to survive treatment and go on to re-infect other parts of the tissue. Antibiotic resistance is a pressing global challenge, and recurrent urinary tract infections are a significant contributor. The bladder-chip presented here could further our understanding of how these bacterial infections develop in vivo and how good antibiotics are at removing them. This could help researchers identify the best dosing and treatment strategies, as well as provide a platform for rapidly testing new antibiotic drugs and other therapies.


Asunto(s)
Técnicas Bacteriológicas/instrumentación , Dispositivos Laboratorio en un Chip , Vejiga Urinaria/irrigación sanguínea , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena , Línea Celular Tumoral , Técnicas de Cocultivo , Células Endoteliales/fisiología , Humanos , Neutrófilos/fisiología
11.
Antibiotics (Basel) ; 10(7)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209966

RESUMEN

To reveal rare phenotypes in bacterial populations, conventional microbiology tools should be advanced to generate rapid, quantitative, accurate, and high-throughput data. The main drawbacks of widely used traditional methods for antibiotic studies include low sampling rate and averaging data for population measurements. To overcome these limitations, microfluidic-microscopy systems have great promise to produce quantitative single-cell data with high sampling rates. Using Mycobacterium smegmatis cells, we applied both conventional assays and a microfluidic-microscopy method to reveal the antibiotic tolerance mechanisms of wild-type and msm2570::Tn mutant cells. Our results revealed that the enhanced antibiotic tolerance mechanism of the msm2570::Tn mutant was due to the low number of lysed cells during the antibiotic exposure compared to wild-type cells. This is the first study to characterize the antibiotic tolerance phenotype of the msm2570::Tn mutant, which has a transposon insertion in the msm2570 gene-encoding a putative xanthine/uracil permease, which functions in the uptake of nitrogen compounds during nitrogen limitation. The experimental results indicate that the msm2570::Tn mutant can be further interrogated to reveal antibiotic killing mechanisms, in particular, antibiotics that target cell wall integrity.

12.
Cell Rep ; 36(3): 109351, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34289360

RESUMEN

Recurrence of uropathogenic Escherichia coli (UPEC) infections has been attributed to reactivation of quiescent intracellular reservoirs (QIRs) in deep layers of the bladder wall. QIRs are thought to arise late during infection following dispersal of bacteria from intracellular bacterial communities (IBCs) in superficial umbrella cells. Here, we track the formation of QIR-like bacteria in a bladder organoid model that recapitulates the stratified uroepithelium within a volume suitable for high-resolution live-cell imaging. Bacteria injected into the organoid lumen enter umbrella-like cells and proliferate to form IBC-like bodies. In parallel, single bacteria penetrate deeper layers of the organoid wall, where they localize within or between uroepithelial cells. These "solitary" bacteria evade killing by antibiotics and neutrophils and are morphologically distinct from bacteria in IBCs. We conclude that bacteria with QIR-like properties may arise at early stages of infection, independent of IBC formation and rupture.


Asunto(s)
Antibacterianos/farmacología , Modelos Biológicos , Neutrófilos/patología , Organoides/microbiología , Vejiga Urinaria/microbiología , Escherichia coli Uropatógena/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Femenino , Humanos , Imagenología Tridimensional , Ratones Endogámicos C57BL , Viabilidad Microbiana/efectos de los fármacos , Movimiento , Neutrófilos/efectos de los fármacos , Organoides/efectos de los fármacos , Organoides/ultraestructura , Vejiga Urinaria/patología , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/crecimiento & desarrollo , Escherichia coli Uropatógena/ultraestructura
13.
EMBO Rep ; 22(6): e52744, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33908688

RESUMEN

Severe cases of SARS-CoV-2 infection are characterized by hypercoagulopathies and systemic endotheliitis of the lung microvasculature. The dynamics of vascular damage, and whether it is a direct consequence of endothelial infection or an indirect consequence of an immune cell-mediated cytokine storm remain unknown. Using a vascularized lung-on-chip model, we find that infection of alveolar epithelial cells leads to limited apical release of virions, consistent with reports of monoculture infection. However, viral RNA and proteins are rapidly detected in underlying endothelial cells, which are themselves refractory to apical infection in monocultures. Although endothelial infection is unproductive, it leads to the formation of cell clusters with low CD31 expression, a progressive loss of barrier integrity and a pro-coagulatory microenvironment. Viral RNA persists in individual cells generating an inflammatory response, which is transient in epithelial cells but persistent in endothelial cells and typified by IL-6 secretion even in the absence of immune cells. Inhibition of IL-6 signalling with tocilizumab reduces but does not prevent loss of barrier integrity. SARS-CoV-2-mediated endothelial cell damage thus occurs independently of cytokine storm.


Asunto(s)
COVID-19 , SARS-CoV-2 , Síndrome de Liberación de Citoquinas , Células Endoteliales , Humanos , Pulmón
14.
J Bacteriol ; 203(10)2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33468595

RESUMEN

Mycobacteria have unique cell envelopes, surface properties, and growth dynamics, which all play a part in the ability of these important pathogens to infect, evade host immunity, disseminate, and resist antibiotic challenges. Recent atomic force microscopy (AFM) studies have brought new insights into the nanometer-scale ultrastructural, adhesive, and mechanical properties of mycobacteria. The molecular forces with which mycobacterial adhesins bind to host factors, like heparin and fibronectin, and the hydrophobic properties of the mycomembrane have been unraveled by AFM force spectroscopy studies. Real-time correlative AFM and fluorescence imaging have delineated a complex interplay between surface ultrastructure, tensile stresses within the cell envelope, and cellular processes leading to division. The unique capabilities of AFM, which include subdiffraction-limit topographic imaging and piconewton force sensitivity, have great potential to resolve important questions that remain unanswered on the molecular interactions, surface properties, and growth dynamics of this important class of pathogens.


Asunto(s)
Membrana Celular/ultraestructura , Mycobacterium/ultraestructura , Adhesinas Bacterianas/metabolismo , Antibacterianos/farmacología , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Interacciones Hidrofóbicas e Hidrofílicas , Lípidos de la Membrana/química , Lípidos de la Membrana/fisiología , Microscopía de Fuerza Atómica , Mycobacterium/química , Mycobacterium/crecimiento & desarrollo , Mycobacterium/fisiología , Propiedades de Superficie
15.
Curr Opin Microbiol ; 59: 72-78, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33049596

RESUMEN

The sum of all of the interactions between single bacteria and host cells determines if an infection is cleared, controlled, or progresses at the whole host-organism level. These individual interactions have independent trajectories defined by diverse and dynamic host-cell and bacterial responses. Focusing on Mycobacterium tuberculosis infection, we discuss how advances in single-cell technologies allow investigation of heterogeneity in host-pathogen interactions and how different layers of heterogeneity in the host affect disease outcome. At late stages of infection, many single interactions co-exist and different outcomes depend on inter-granuloma and intra-granuloma heterogeneity. However, during bottleneck events involving small numbers of bacteria, random events, such as chance interactions with more or less permissive host cells, play a decisive role and may explain why some exposed individuals never develop the disease.


Asunto(s)
Comunicación Celular , Interacciones Huésped-Patógeno , Tuberculosis , Comunicación Celular/inmunología , Granuloma/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Tuberculosis Latente/inmunología , Tuberculosis Latente/fisiopatología , Mycobacterium tuberculosis , Tuberculosis/inmunología , Tuberculosis/fisiopatología
16.
Biofabrication ; 13(2)2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33186924

RESUMEN

Despite its simplicity, which makes it the most commonly used bioprinting method today, extrusion-based bioprinting suffers from its inability to reproduce the complex tissue architecture found in organs. Generally, this printing method allows for the dispensing of solutions of a predefined cell concentration through a rudimentary needle. Moreover, to avoid cell lysis in the dispensing needle, which is detrimental to the viability of the printed tissue, as well as cell loss in dead volumes of tubing, thereby increasing the cost of printing tissue, a common strategy has been to print with cell concentrations much lower in comparison to the concentrations found in living tissues. As a result, cell-to-cell distance is increased in the dispensed samples impairing communication through cytokines. Here, we present a microfluidic-based print head capable of modulating the printed cell concentration in real-time. This device allows bioprinting at high cell concentrations by concentrating and dispensing fibroblasts at concentrations up to 10 million cells∙mL-1. We also demonstrate that this device can be used to print bladder organoids. As the cell seeding concentration is of major importance for organogenesis in 3D culture, organoid printing allows the user to standardize the process of organoid formation and achieve more reliable and reproducible results.


Asunto(s)
Bioimpresión , Fibroblastos , Microfluídica , Organoides , Impresión Tridimensional , Ingeniería de Tejidos , Andamios del Tejido
17.
Elife ; 92020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33228849

RESUMEN

We establish a murine lung-on-chip infection model and use time-lapse imaging to reveal the dynamics of host-Mycobacterium tuberculosis interactions at an air-liquid interface with a spatiotemporal resolution unattainable in animal models and to probe the direct role of pulmonary surfactant in early infection. Surfactant deficiency results in rapid and uncontrolled bacterial growth in both macrophages and alveolar epithelial cells. In contrast, under normal surfactant levels, a significant fraction of intracellular bacteria are non-growing. The surfactant-deficient phenotype is rescued by exogenous addition of surfactant replacement formulations, which have no effect on bacterial viability in the absence of host cells. Surfactant partially removes virulence-associated lipids and proteins from the bacterial cell surface. Consistent with this mechanism, the attenuation of bacteria lacking the ESX-1 secretion system is independent of surfactant levels. These findings may partly explain why smokers and elderly persons with compromised surfactant function are at increased risk of developing active tuberculosis.


Tuberculosis is a contagious respiratory disease caused by the bacterium Mycobacterium tuberculosis. Droplets in the air carry these bacteria deep into the lungs, where they cling onto and infect lung cells. Only small droplets, holding one or two bacteria, can reach the right cells, which means that just a couple of bacterial cells can trigger an infection. But people respond differently to the bacteria: some develop active and fatal forms of tuberculosis, while many show no signs of infection. With no effective tuberculosis vaccine for adults, understanding why individuals respond differently to Mycobacterium tuberculosis may help develop treatments. Different responses to Mycobacterium tuberculosis may stem from the earliest stages of infection, but these stages are difficult to study. For one thing, tracking the movements of the few bacterial cells that initiate infection is tricky. For another, studying the molecules, called 'surfactants', that the lungs produce to protect themselves from tuberculosis can prove difficult because these molecules are necessary for the lungs to inflate and deflate normally. Normally, the role of a molecule can be studied by genetically modifying an animal so it does not produce the molecule in question, which provides information as to its potential roles. Unfortunately, due to the role of surfactants in normal breathing, animals lacking them die. Therefore, to reveal the role of some of surfactants in tuberculosis, Thacker et al. used 'lung-on-chip' technology. The 'chip' (a transparent device made of a polymer compatible with biological tissues) is coated with layers of cells and has channels to simulate air and blood flow. To see what effects surfactants have on M. tuberculosis bacteria, Thacker et al. altered the levels of surfactants produced by the cells on the lung-on-chip device. Two types of mouse cells were grown on the chip: lung cells and immune cells. When cells lacked surfactants, bacteria grew rapidly on both lung and immune cells, but when surfactants were present bacteria grew much slower on both cell types, or did not grow at all. Further probing showed that the surfactants pulled out proteins and fats on the surface of M. tuberculosis that help the bacteria to infect their host, highlighting the protective role of surfactants in tuberculosis. These findings lay the foundations for a system to study respiratory infections without using animals. This will allow scientists to study the early stages of Mycobacterium tuberculosis infection, which is crucial for finding ways to manage tuberculosis.


Asunto(s)
Células Epiteliales Alveolares/microbiología , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/instrumentación , Mycobacterium tuberculosis/crecimiento & desarrollo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Tuberculosis Pulmonar/microbiología , Células Epiteliales Alveolares/metabolismo , Animales , Carga Bacteriana , Proteínas Bacterianas/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Viabilidad Microbiana , Microscopía por Video , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Proteínas Asociadas a Surfactante Pulmonar/genética , Factores de Tiempo , Imagen de Lapso de Tiempo , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/metabolismo , Virulencia
18.
Adv Biosyst ; 4(2): e1900103, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32293136

RESUMEN

Three models of division control are proposed to achieve cell size homeostasis: sizer, timer, and adder. However, few published studies of division control take into account the dynamics of single-cell growth and most assume that single-cell growth is exponential. Here, computational simulations considering exponential, linear, and bilinear growth models are performed. These simulations confirm that a timer division control model alone cannot lead to size homeostasis if the single-cell growth model is exponential. Furthermore, timer and adder division control models cannot be distinguished if the single-cell growth model is linear. Models of division control cannot be easily differentiated by analysis of average cell behavior because the birth sizes of the majority of cells are close to the population average. However, the differences between division control models are amplified in outlier cells whose birth size is far from the average. A method is introduced for vector field analysis of the speed of convergence of outlier lineages toward the steady-state birth size, which can help to distinguish between division control models and single-cell growth models.


Asunto(s)
División Celular/fisiología , Tamaño de la Célula , Simulación por Computador , Modelos Biológicos , Proliferación Celular/fisiología , Homeostasis/fisiología , Análisis de la Célula Individual
19.
Nat Phys ; 16(1): 57-62, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31921326

RESUMEN

Mechanisms to control cell division are essential for cell proliferation and survival 1. Bacterial cell growth and division require the coordinated activity of peptidoglycan synthases and hydrolytic enzymes 2-4 to maintain mechanical integrity of the cell wall 5. Recent studies suggest that cell separation is governed by mechanical forces 6,7. How mechanical forces interact with molecular mechanisms to control bacterial cell division in space and time is poorly understood. Here, we use a combination of atomic force microscope (AFM) imaging, nanomechanical mapping, and nanomanipulation to show that enzymatic activity and mechanical forces serve overlapping and essential roles in mycobacterial cell division. We find that mechanical stress gradually accumulates in the cell wall concentrated at the future division site, culminating in rapid (millisecond) cleavage of nascent sibling cells. Inhibiting cell wall hydrolysis delays cleavage; conversely, locally increasing cell wall stress causes instantaneous and premature cleavage. Cells deficient in peptidoglycan hydrolytic activity fail to locally decrease their cell wall strength and undergo natural cleavage, instead forming chains of non-growing cells. Cleavage of these cells can be mechanically induced by local application of stress with AFM. These findings establish a direct link between actively controlled molecular mechanisms and passively controlled mechanical forces in bacterial cell division.

20.
Nat Commun ; 11(1): 452, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31974342

RESUMEN

Mycobacteria grow by inserting new cell wall material in discrete zones at the cell poles. This pattern implies that polar growth zones must be assembled de novo at each division, but the mechanisms that control the initiation of new pole growth are unknown. Here, we combine time-lapse optical and atomic force microscopy to measure single-cell pole growth in mycobacteria with nanometer-scale precision. We show that single-cell growth is biphasic due to a lag phase of variable duration before the new pole transitions from slow to fast growth. This transition and cell division are independent events. The difference between the lag and interdivision times determines the degree of single-cell growth asymmetry, which is high in fast-growing species and low in slow-growing species. We propose a biphasic growth model that is distinct from previous unipolar and bipolar models and resembles "new end take off" (NETO) dynamics of polar growth in fission yeast.


Asunto(s)
Modelos Biológicos , Mycobacterium/citología , Mycobacterium/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , División Celular , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía de Fuerza Atómica , Mycobacterium/genética , Análisis Espacio-Temporal , Imagen de Lapso de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA