Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Microbiol Mol Biol Rev ; 87(4): e0017422, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37962348

ABSTRACT

SUMMARYAntibiotic persistence, or the ability of small subsets of bacteria to survive prolonged antibiotic treatment, is an underappreciated cause of antibiotic treatment failure. Over the past decade, researchers have discovered multiple different stress responses and mechanisms that can promote antibiotic persistence. However, many of these studies have been completed in culture-based systems that fail to truly replicate the complexities of the host environment, and it is unclear whether the mechanisms defined in in vitro studies are applicable during host infection. In this review, we focus our discussion on recent studies that utilize a mixture of ex vivo culture systems and animal models to understand what stressors in the host environment are important for inducing antibiotic persistence. Different host stressors are involved depending on the anatomical niche the bacteria reside in and whether the host immune system is primed to generate a more robust response against bacteria, which can result in differing downstream effects on antibiotic susceptibility. Bacterial pathogens can also utilize specific strategies to reprogram their metabolism, which is vital for transitioning into an antibiotic-persistent state within host tissues. Importantly, we highlight that more attention is needed to establish guidelines for in vivo work on antibiotic persistence, particularly when identifying antibiotic-persistent subpopulations and distinguishing these phenotypes from antibiotic tolerance. Studying antibiotic persistence in the context of the host environment will be crucial for developing tools and strategies to target antibiotic-persistent bacteria and increase the efficacy of antibiotic treatment.


Subject(s)
Anti-Bacterial Agents , Bacterial Infections , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Bacterial Infections/drug therapy , Bacterial Infections/microbiology
2.
STAR Protoc ; 3(4): 101760, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36219561

ABSTRACT

Yersinia pseudotuberculosis (Yptb) is a bacterial pathogen that causes foodborne illness. Defense against the host antimicrobial gas, nitric oxide (NO), by the bacterial NO-detoxifying gene, hmp, promotes Yptb replication in mouse models of infection. Here, we detail the use of fluorescent signals as readouts for NO exposure within individual cells and subsequent detection of heterogeneity within a population, using single-cell imaging and analysis. This protocol quantifies NO exposure in culture, without capturing the full complexity of the host environment. For complete details on the use and execution of this protocol, please refer to Patel et al. (2021).


Subject(s)
Yersinia pseudotuberculosis Infections , Yersinia pseudotuberculosis , Animals , Mice , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/microbiology , Nitric Oxide
3.
Infect Immun ; 90(8): e0016722, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35862700

ABSTRACT

Fluorescence dilution approaches can detect bacterial cell division events and can detect if there are differential rates of cell division across individual cells within a population. This approach typically involves inducing expression of a fluorescent protein and then tracking partitioning of fluorescence into daughter cells. However, fluorescence can be diluted very quickly within a rapidly replicating population, such as pathogenic bacterial populations replicating within host tissues. To overcome this limitation, we have generated two revTetR reporter constructs, where either mCherry or yellow fluorescent protein (YFP) is constitutively expressed and repressed by addition of tetracyclines, resulting in fluorescence dilution within defined time frames. We show that fluorescent signals are diluted in replicating populations and that signal accumulates in growth-inhibited populations, including during nitric oxide (NO) exposure. Furthermore, we show that tetracyclines can be delivered to the mouse spleen during Yersinia pseudotuberculosis infection and defined a drug concentration that results in even exposure of cells to tetracyclines. We then used this system to visualize bacterial cell division within defined time frames postinfection. revTetR-mCherry allowed us to detect slow-growing cells in response to NO in culture; however, this strain had a growth defect within mouse tissues, which complicated results. To address this issue, we constructed revTetR-YFP using the less toxic YFP and showed that heightened NO exposure correlated with heightened YFP signal, indicating decreased cell division rates within this subpopulation in vivo. This revTetR reporter will provide a critical tool for future studies to identify and isolate slowly replicating bacterial subpopulations from host tissues.


Subject(s)
Yersinia pseudotuberculosis Infections , Yersinia pseudotuberculosis , Animals , Cell Division , Mice , Nitric Oxide/metabolism , Spleen/microbiology , Tetracyclines , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/microbiology
4.
PLoS Pathog ; 18(5): e1010556, 2022 05.
Article in English | MEDLINE | ID: mdl-35576231

ABSTRACT

Antibiotic tolerance is typically associated with a phenotypic change within a bacterial population, resulting in a transient decrease in antibiotic susceptibility that can contribute to treatment failure and recurrent infections. Although tolerant cells may emerge prior to treatment, the stress of prolonged antibiotic exposure can also promote tolerance. Here, we sought to determine how Yersinia pseudotuberculosis responds to doxycycline exposure, to then verify if these gene expression changes could promote doxycycline tolerance in culture and in our mouse model of infection. Only four genes were differentially regulated in response to a physiologically-relevant dose of doxycycline: osmB and ompF were upregulated, tusB and cnfy were downregulated; differential expression also occurred during doxycycline treatment in the mouse. ompF, tusB and cnfy were also differentially regulated in response to chloramphenicol, indicating these could be general responses to ribosomal inhibition. cnfy has previously been associated with persistence and was not a major focus here. We found deletion of the OmpF porin resulted in increased antibiotic accumulation, suggesting expression may promote diffusion of doxycycline out of the cell, while OsmB lipoprotein had a minor impact on antibiotic permeability. Overexpression of tusB significantly impaired bacterial survival in culture and in the mouse, suggesting that tRNA modification by tusB, and the resulting impacts on translational machinery, promotes survival during treatment with an antibiotic classically viewed as bacteriostatic. We believe this may be the first observation of bactericidal activity of doxycycline under physiological conditions, which was revealed by reversing tusB downregulation.


Subject(s)
Yersinia pseudotuberculosis , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Doxycycline/metabolism , Doxycycline/pharmacology , Mice , Permeability , RNA, Transfer/metabolism , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/metabolism
5.
Proc Natl Acad Sci U S A ; 117(1): 629-634, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31871194

ABSTRACT

Antibiotic resistance in bacteria is typically conferred by proteins that function as efflux pumps or enzymes that modify either the drug or the antibiotic target. Here we report an unusual mechanism of resistance to macrolide-lincosamide antibiotics mediated by mycobacterial HflX, a conserved ribosome-associated GTPase. We show that deletion of the hflX gene in the pathogenic Mycobacterium abscessus, as well as the nonpathogenic Mycobacterium smegmatis, results in hypersensitivity to the macrolide-lincosamide class of antibiotics. Importantly, the level of resistance provided by Mab_hflX is equivalent to that conferred by erm41, implying that hflX constitutes a significant resistance determinant in M. abscessus We demonstrate that mycobacterial HflX associates with the 50S ribosomal subunits in vivo and can dissociate purified 70S ribosomes in vitro, independent of GTP hydrolysis. The absence of HflX in a ΔMs_hflX strain also results in a significant accumulation of 70S ribosomes upon erythromycin exposure. Finally, a deletion of either the N-terminal or the C-terminal domain of HflX abrogates ribosome splitting and concomitantly abolishes the ability of mutant proteins to mediate antibiotic tolerance. Together, our results suggest a mechanism of macrolide-lincosamide resistance in which the mycobacterial HflX dissociates antibiotic-stalled ribosomes and rescues the bound mRNA. Given the widespread presence of hflX genes, we anticipate this as a generalized mechanism of macrolide resistance used by several bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Drug Resistance, Bacterial/genetics , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium abscessus/physiology , Ribosome Subunits, Large, Bacterial/metabolism , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Humans , Lincosamides/pharmacology , Lincosamides/therapeutic use , Macrolides/pharmacology , Macrolides/therapeutic use , Mutation , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium abscessus/drug effects , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/physiology , Protein Biosynthesis/drug effects , Protein Domains/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...